1
|
Liu Y, Zhong L, Sun Z, Feng Y, Ding Q, Zhang Y. N-3 Fatty Acid Supplementation in Mothers and Infants for Childhood Psychomotor and Cognitive Development: An Updated Systematic Review and Meta-Analysis. MATERNAL & CHILD NUTRITION 2025; 21:e13767. [PMID: 39609259 PMCID: PMC11956035 DOI: 10.1111/mcn.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/30/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
Long-chain n-3 polyunsaturated fatty acid (PUFA) consumption in maternal and infants has been positively associated with cognitive and visual development. Tails even meta-analysis showed mixed results. To evaluate the effects of maternal and infant n-3 PUFA supplementation on childhood psychomotor and cognitive development, PubMed, Embase, the Cochrane Library, PsycINFO and clinicaltrials.gov were searched. Randomized controlled trials were included to evaluate the effect on child cognitive and psychomotor outcomes of n-3 PUFA supplementation in mothers or infants (age ≤ 2 years). Findings were pooled with mean differences (MD) with 95% confidence intervals (95% CIs). Heterogeneity was explored using I2 and subgroup analyses, stratified for maternal (pregnancy and/or lactation) and infant (preterm infant and term infant). We identified 47 articles, with 14 trials on mothers and 33 on infants. Pooled results showed that infants' mental development index (MDI) increased with n-3 PUFA supplementation (MD = 2.91, 95% CI: 1.32-4.51, I2 = 65.1%). Subgroup analysis of MDI also demonstrated a benefit in preterm infants (MD = 4.16, 95% CI: 1.40-6.93, I2 = 49.5%) and term infants (MD = 2.28, 95% CI: 0.27-4.29, I2 = 70.1%). No significant association was found in subgroup analyses of supplementation to mothers during pregnancy or lactation period. Supplementation did not increase the psychomotor development index (PDI) in the mother or infant group. Language composite score increased for infants whose mothers accepted supplementation in pregnancy or breastfeeding (MD = 8.57, 95% CI: 5.09-12.04, I2 = 70.2%). The cognitive composite score did not improve in any subgroup. Intelligence Quotient (IQ) increased in the infants' group with n-3 PUFA supplementation (MD = 2.54, 95% CI: 0.45-4.63, I2 = 66.0%). Furthermore, IQ in term infants also improved (MD = 2.91, 95% CI: 0.24-5.57, I2 = 69.2%). The funnel plot and Egger's test confirmed no publication bias in any endpoints. Supplementation with n-3 PUFA during pregnancy or breastfeeding in mothers has increased language abilities. Furthermore, direct supplementation in term infants can improve intelligence in later childhood. However, insufficient evidence supports the claim that supplementation improves cognitive abilities.
Collapse
Affiliation(s)
- Yingyu Liu
- Division of Surveillance and EvaluationChinese Center for Health EducationBeijingChina
- Department of Epidemiology and BiostatisticsSchool of Public HealthJilin UniversityChangchunChina
| | - Lijun Zhong
- Department of Cancer Prevention and Control, Sichuan Clinical Research Center for CancerSichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Zhouyang Sun
- Department of Epidemiology and BiostatisticsSchool of Public HealthJilin UniversityChangchunChina
| | - Yuan Feng
- Department of Epidemiology and BiostatisticsSchool of Public HealthJilin UniversityChangchunChina
| | - Qianlu Ding
- Department of Epidemiology and BiostatisticsSchool of Public HealthJilin UniversityChangchunChina
| | - Yujian Zhang
- Department of Cancer Prevention and Control, Sichuan Clinical Research Center for CancerSichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
2
|
Peh HY, Chen J. Pro-resolving lipid mediators and therapeutic innovations in resolution of inflammation. Pharmacol Ther 2025; 265:108753. [PMID: 39566561 DOI: 10.1016/j.pharmthera.2024.108753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
This review summarizes findings presented at the 19th World Congress of Basic & Clinical Pharmacology 2023 (Glasgow, Scotland, July 3rd to 7th, 2023) from 8 speakers in the field of resolution of inflammation, resolution pharmacology and resolution biology. It is now accepted that the acute inflammatory response is protective to defend the host against infection or tissue injury. Acute inflammation is self-limited and programmed to be limited in space and time: this is achieved through endogenous resolution processes that ensure return to homeostasis. Resolution is brought about by agonist mediators that include specialized pro-resolving lipid mediators (SPMs) and pro-resolving proteins and peptides such as annexin A1 and angiotensin-(1-7), all acting to initiate anti-inflammatory and pro-resolving processes. If the inflammatory reaction remains unchecked through dysfunctional resolution mechanism, it can become chronic and contribute to a plethora of human diseases, including respiratory, cardiovascular, metabolic, allergic diseases, and arthritis. Herein, we discuss how non-resolving inflammation plays a role in the pathogenesis of these diseases. In addition to SPMs, we highlight the discovery, biosynthesis, biofunctions, and latest research updates on innovative therapeutics (including annexin-A1 peptide-mimetic RTP-026, small molecule FPR2 agonist BM-986235/LAR-1219, biased agonist for FPR1/FPR2 Cmpd17b, lipoxin mimetics AT-01-KG and AT-02-CT, melanocortin receptor agonist AP1189, gold nanoparticles, angiotensin-(1-7), and CD300a) that can promote resolution of inflammation directly or through modulation of SPMs production. Drug development strategies based on the biology of the resolution of inflammation can offer novel therapeutic means and/or add-on therapies for the treatment of chronic diseases.
Collapse
Affiliation(s)
- Hong Yong Peh
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Pharmacology, Singapore; Immunology Programme and Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
3
|
Ottolini KM, Ngwa J, Basu SK, Kapse K, Liggett M, Murnick J, Limperopoulos C, Andescavage N. Brain development using a multicomponent intravenous lipid emulsion in preterm infants. BMC Pediatr 2024; 24:847. [PMID: 39736580 DOI: 10.1186/s12887-024-05330-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/12/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Intravenous lipid emulsions are an essential component of nutritional support for very preterm infants. Many neonatal intensive care units have transitioned from traditional soybean oil-only to fish oil-containing multicomponent lipid emulsions, but the neurodevelopmental implications have not been well-explored. The primary aim of this study was to assess extrauterine third trimester brain growth in very preterm infants supported with soybean oil-only compared to fish-oil containing multicomponent lipid emulsions; white matter development and neurobehavioral regulation at term were also investigated. METHODS Human milk-fed very preterm infants (born less than or equal to 32 weeks' gestation) receiving either soybean oil-only (before 2019) or multicomponent (after 2019) lipid emulsions underwent quantitative brain MRI (volumetric growth and white matter development) and neurodevelopmental assessment (Neonatal Intensive Care Unit Network Neurobehavioral Scale) at term-equivalent age. Analyses were adjusted for age at birth and term assessments, as well as clinically significant covariates. RESULTS 92 infants (61 soybean, 31 multicomponent) were included (mean [SD] birth gestational age: 27.3 [2.3] weeks). Soybean oil-only infants demonstrated smaller brainstem volumes (β [95% CI] = -0.5 [-0.8,-0.1], p = .007); additionally less mature white matter development (mean diffusivity [MD, mm2/second x10- 3] and fractional anisotropy [FA]) in the corpus callosum (MD genu: β = 0.10 [0.01, 0.20], p = .04; splenium: β = 0.14 [0.04, 0.24], p = .006), posterior limbs of internal capsule (MD right (R): β = 0.05 [0.02, 0.08], p = .004, left (L): β = 0.04 [0.01, 0.08], p = .01; FA R: β = -0.03 [-0.06, -0.00], p = .03), and brainstem (FA R: β = 0.07 [0.04, 0.10], p < .001, L: β = 0.05 [0.02, 0.09], p = .002); and lower quality of movement (β = -0.54 [-0.97, -0.11], p = .02) and higher state-related stress (β = 1.41 [0.14, 2.83], p = .04). CONCLUSIONS Very preterm infants supported with a fish-oil containing multicomponent compared to soybean oil-only lipid emulsion demonstrated improved regional brain growth, as well as evidence of enhanced white matter microstructural organization and neurobehavioral regulation, at term corrected age. TRIAL REGISTRATION Clinical trial number: Not applicable.
Collapse
Affiliation(s)
- Katherine M Ottolini
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
- Division of Neonatology, Children's National Hospital, Washington, DC, 20010, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Julius Ngwa
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sudeepta K Basu
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
- Division of Neonatology, Children's National Hospital, Washington, DC, 20010, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Kushal Kapse
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
| | - Melissa Liggett
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
| | - Jonathan Murnick
- Division of Diagnostic Imaging and Radiology, Children's National Hospital, Washington, DC, USA
- Department of Radiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Radiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Nickie Andescavage
- Developing Brain Institute, Children's National Hospital, 111 Michigan Avenue, NW, Washington, DC, USA.
- Division of Neonatology, Children's National Hospital, Washington, DC, 20010, USA.
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
4
|
Fang X, Lee S, Rayalam S, Park HJ. Docosahexaenoic acid supplementation and infant brain development: role of gut microbiome. Nutr Res 2024; 131:1-13. [PMID: 39342808 DOI: 10.1016/j.nutres.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Perinatal stage represents a critical period for brain development. Docosahexaenoic acid (DHA) is a ω-3 polyunsaturated fatty acid preferentially accumulated in the brain that may benefit neurodevelopment. Microbial colonization and maturation parallel with the rapid development of infant metabolic and brain function that may influence the effects of DHA on neurological development. This review aims to summarize the current literature on the mediating effects of DHA on brain and gut microbiome development and attempts to reevaluate the efficacy of DHA from a gut microbiome-mediated perspective. Specifically, the regulatory roles of DHA on hypothalamic-pituitary-adrenal axis, inflammation, and neuroactive mediators may be partly moderated through gut microbiome. Consideration of the gut microbiome and gut-brain communication, when evaluating the efficacy of DHA, may provide new insights in better understanding the mechanisms of DHA and impart advantages to future development of nutritional therapy based on the nutrient-microbiome interaction.
Collapse
Affiliation(s)
- Xi Fang
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA
| | - Soon Lee
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA
| | - Srujana Rayalam
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA
| | - Hea Jin Park
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA.
| |
Collapse
|
5
|
Luo S, Zhang J, Sun J, Zhao T, Deng J, Yang H. Future development trend of food-borne delivery systems of functional substances for precision nutrition. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 112:385-433. [PMID: 39218507 DOI: 10.1016/bs.afnr.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
BACKGROUND Precision nutrition, a personalized nutritional supplementation model, is widely acknowledged for its significant impact on human health. Nevertheless, challenges persist in the advancement of precision nutrition, including consumer dietary behaviors, nutrient absorption, and utilization. Thus, the exploration of effective strategies to enhance the efficacy of precision nutrition and maximize its potential benefits in dietary interventions and disease management is imperative. SCOPE AND APPROACH The primary objective of this comprehensive review is to synthesize and assess the latest technical approaches and future prospects for achieving precision nutrition, while also addressing the existing constraints in this field. The role of delivery systems is pivotal in the realization of precision nutrition goals. This paper outlines the potential applications of delivery systems in precision nutrition and highlights key considerations for their design and implementation. Additionally, the review offers insights into the evolving trends in delivery systems for precision nutrition, particularly in the realms of nutritional fortification, specialized diets, and disease prevention. KEY FINDINGS AND CONCLUSIONS By leveraging computer data collection, omics, and metabolomics analyses, this review scrutinizes the lifestyles, dietary patterns, and health statuses of diverse organisms. Subsequently, tailored nutrient supplementation programs are devised based on individual organism profiles. The utilization of delivery systems enhances the bioavailability of functional compounds and enables targeted delivery to specific body regions, thereby catering to the distinct nutritional requirements and disease prevention needs of consumers, with a particular emphasis on special populations and dietary preferences.
Collapse
Affiliation(s)
- Shuwei Luo
- College of Food Science and Nutritional and Engineering, China Agricultural University, Beijing, P.R. China
| | - Juntao Zhang
- College of Food Science and Nutritional and Engineering, China Agricultural University, Beijing, P.R. China
| | - Jing Sun
- College of Food Science and Nutritional and Engineering, China Agricultural University, Beijing, P.R. China
| | - Tong Zhao
- College of Food Science and Nutritional and Engineering, China Agricultural University, Beijing, P.R. China
| | - Jianjun Deng
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P.R. China
| | - Haixia Yang
- College of Food Science and Nutritional and Engineering, China Agricultural University, Beijing, P.R. China.
| |
Collapse
|
6
|
Xu Q, Wang S, Zou Q, Chen W, Lan D, Wang Y. Streamlined enzymatic synthesis of human milk fat substitutes. Food Chem 2024; 443:138476. [PMID: 38306908 DOI: 10.1016/j.foodchem.2024.138476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 02/04/2024]
Abstract
The positional distribution of palmitic acid (PA) in human milk fat substitutes (HMFSs) plays a pivotal role in mimicking the nutritional profile of human milk fat for nourishing non-breastfed infants. This study innovatively introduced a streamlined enzymatic process for preparing HMFSs rich in sn-2 PA using palm stearin, a PA-rich source without the necessity for positional distribution of PA. The initial step involved enhancing the sn-2 PA concentration through enzymatic interesterification using Lipase UM1, which exhibited superior catalytic efficiency than Novozym 435. This process increased the sn-2 PA level from 40.98 % to 64.51 %. Subsequently, acidolysis was employed to reduce PA levels by replacing PA at sn-1,3 positions using sn-1,3-regioselective lipases. The PA content decreased from 60.64 % to 26.73 %, simultaneously raising the relative sn-2 PA concentration to 71.57 %, meeting the benchmarks for HMFSs. This study establishes a robust conceptual framework for the prospective industrial synthesis of HMFSs.
Collapse
Affiliation(s)
- Qingqing Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Shushu Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Qian Zou
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Wen Chen
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Dongming Lan
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China.
| | - Yonghua Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, PR China; Guangdong Yue-shan Special Nutrition Technology Co. Ltd., Foshan 528000, PR China.
| |
Collapse
|
7
|
Smolińska K, Szopa A, Sobczyński J, Serefko A, Dobrowolski P. Nutritional Quality Implications: Exploring the Impact of a Fatty Acid-Rich Diet on Central Nervous System Development. Nutrients 2024; 16:1093. [PMID: 38613126 PMCID: PMC11013435 DOI: 10.3390/nu16071093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024] Open
Abstract
Given the comprehensive examination of the role of fatty acid-rich diets in central nervous system development in children, this study bridges significant gaps in the understanding of dietary effects on neurodevelopment. It delves into the essential functions of fatty acids in neurodevelopment, including their contributions to neuronal membrane formation, neuroinflammatory modulation, neurogenesis, and synaptic plasticity. Despite the acknowledged importance of these nutrients, this review reveals a lack of comprehensive synthesis in current research, particularly regarding the broader spectrum of fatty acids and their optimal levels throughout childhood. By consolidating the existing knowledge and highlighting critical research gaps, such as the effects of fatty acid metabolism on neurodevelopmental disorders and the need for age-specific dietary guidelines, this study sets a foundation for future studies. This underscores the potential of nutritional strategies to significantly influence neurodevelopmental trajectories, advocating an enriched academic and clinical understanding that can inform dietary recommendations and interventions aimed at optimizing neurological health from infancy.
Collapse
Affiliation(s)
- Katarzyna Smolińska
- Chronic Wounds Laboratory, Medical University of Lublin, Chodźki St. 7, 20-093 Lublin, Poland;
| | - Aleksandra Szopa
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Jan Sobczyński
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Anna Serefko
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Maria Curie Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland
| |
Collapse
|
8
|
Hu R, Xu J, Hua Y, Li Y, Li J. Could early life DHA supplementation benefit neurodevelopment? A systematic review and meta-analysis. Front Neurol 2024; 15:1295788. [PMID: 38645744 PMCID: PMC11032049 DOI: 10.3389/fneur.2024.1295788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Background Docosahexaenoic acid (DHA) plays a crucial role in the growth and functional development of the infant brain. However, the impact of additional DHA supplementation on neurodevelopment in infants remains controversial in randomized controlled trials. In this systematic review and meta-analysis, we aimed to investigate the effects of prenatal and postnatal DHA supplementation on neurodevelopment. Methods We systematically searched the MEDLINE, EMBASE, and Cochrane Library electronic databases using a predefined strategy until 8 February 2024. We extracted relevant study characteristics and outcomes related to the nervous system. Two independent reviewers critically evaluated the included studies to assess their validity and risk of bias. Results A total of 21 studies met our inclusion criteria, one study was removed after quality assessment, and the meta-analysis included 9 randomized controlled trials. The meta-analysis results indicated that there was no statistically significant difference between the DHA supplementation group and the placebo group, as assessed by the Mental Development Index [MDI; mean difference (MD), 0.41; 95% confidence interval (CI), -0.91 to 1.73; p = 0.55]. However, the DHA group had a significantly higher Psychomotor Development Index (PDI) than the placebo group (MD, 1.47; 95% CI, 0.23 to 2.72; p = 0.02). Subgroup analyses based on populations showed that DHA supplementation was superior to placebo for infants in both MDI (language score conversion; MD, 2.05; 95% CI, -0.16 to 4.26; p = 0.07) and PDI (MD, 1.94; 95% CI, 0.23 to 3.65; p = 0.03). Other subgroup analyses indicated no statistical differences between the two groups. The remaining assessments that could not be summarized quantitatively underwent a narrative evaluation. Conclusion Based on the BSID assessments, DHA supplementation in infants may have potential neurodevelopmental benefits. Because the meta-analysis included few high-quality articles and had some limitations, more relevant articles are needed to address the need for separate DHA supplementation in infants, pregnant women, and lactating mothers. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022348100, identifier: CRD42022348100.
Collapse
Affiliation(s)
| | | | | | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinrong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Fan L, Wang X, Szeto IMY, Liu B, Sinclair AJ, Li D. Dietary intake of different ratios of ARA/DHA in early stages and its impact on infant development. Food Funct 2024; 15:3259-3273. [PMID: 38469864 DOI: 10.1039/d3fo04629j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFAs), arachidonic acid (ARA, 20:4n-6) and docosahexaenoic acid (DHA, 22:6n-3) are essential in the development of infants. ARA and DHA from breast milk or infant formula are the main sources of access for infants to meet their physiological and metabolic needs. The ratio of ARA to DHA in breast milk varies among regions and different lactation stages. Different ratios of ARA and DHA mainly from algal oil, animal fat, fish oil, and microbial oil, are added to infant formula in different regions and infant age ranges. Supplementing with appropriate ratios of ARA and DHA during infancy promotes brain, neural, visual, and other development aspects. In this review, we first introduced the current intake status of ARA and DHA in different locations, lactation stages, and age ranges in breast milk and infant formula. Finally, we discussed the effect of different ratios of ARA and DHA on infant development. This review provided a comprehensive research basis for the nutritional research of infants who consume different ratios of ARA and DHA.
Collapse
Affiliation(s)
- Lijiao Fan
- Institute of Nutrition & Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Xincen Wang
- Institute of Nutrition & Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| | | | - Biao Liu
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Andrew J Sinclair
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Notting Hill, VIC 3168, Australia
- Faculty of Health, Deakin University, Burwood, VIC 3152, Australia
| | - Duo Li
- Institute of Nutrition & Health, School of Public Health, Qingdao University, Qingdao 266071, China.
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Notting Hill, VIC 3168, Australia
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Shahabi B, Hernández-Martínez C, Voltas N, Canals J, Arija V. The Maternal Omega-3 Long-Chain Polyunsaturated Fatty Acid Concentration in Early Pregnancy and Infant Neurodevelopment: The ECLIPSES Study. Nutrients 2024; 16:687. [PMID: 38474815 DOI: 10.3390/nu16050687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Omega-3 Long-Chain Polyunsaturated Fatty Acids (n-3 LCPUFAs) play a key role in early neurodevelopment, but evidence from observational and clinical studies remains inconsistent. This study investigates the association between maternal n-3 LCPUFA, Docosahexaenoic Acid (DHA), and eicosapentaenoic acid (EPA) concentrations during pregnancy and infant development functioning at 40 days. This study includes 348 mother-infant pairs. Maternal serum concentrations were assessed in the first and third trimesters alongside sociodemographic, clinical, nutritional, psychological, and obstetrical data. At 40 days, the Bayley Scales of Infant and Toddler Development, Third Edition (BSID-III) was administered. An adjusted analysis revealed that lower first-trimester n-3 LCPUFA and DHA concentrations are associated with better infant motor development. These results underscore the potential significance of the maternal n-3 LCPUFA status in early pregnancy for influencing fetal neurodevelopment. However, the complexity of these associations necessitates further investigation, emphasizing the urgent need for additional studies to comprehensively elucidate the nuanced interplay between the maternal n-3 LCPUFA status and infant neurodevelopment.
Collapse
Affiliation(s)
- Behnaz Shahabi
- Research Group in Nutrition and Mental Health (NUTRISAM), Universitat Rovira i Virgili, 43201 Reus, Spain
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Carmen Hernández-Martínez
- Research Group in Nutrition and Mental Health (NUTRISAM), Universitat Rovira i Virgili, 43201 Reus, Spain
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain
- Research Center for Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Núria Voltas
- Research Group in Nutrition and Mental Health (NUTRISAM), Universitat Rovira i Virgili, 43201 Reus, Spain
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain
- Research Center for Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, 43003 Tarragona, Spain
- Serra Húnter Fellow, Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Josefa Canals
- Research Group in Nutrition and Mental Health (NUTRISAM), Universitat Rovira i Virgili, 43201 Reus, Spain
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain
- Research Center for Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Victoria Arija
- Research Group in Nutrition and Mental Health (NUTRISAM), Universitat Rovira i Virgili, 43201 Reus, Spain
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain
| |
Collapse
|
11
|
Ikeda N, Shepherd E, Makrides M, McPhee AJ, Gibson RA, Gould JF. The effects of parenteral fish oil on neurodevelopment in preterm infants: A narrative review. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102620. [PMID: 38763084 DOI: 10.1016/j.plefa.2024.102620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE This narrative review aimed to summarize studies assessing the effects of parenteral fish oil on neurodevelopment in preterm infants. METHODS PubMed was searched (July 1985 to October 2023). We reviewed randomized controlled trials, and observational studies assessing intravenous lipid emulsion with fish oil in preterm infants (born less than 37 weeks' gestation), that reported long-term neurodevelopmental outcomes. RESULTS We identified four publications relating to three randomized controlled trials in addition to four cohort studies. Study designs and outcomes were heterogenous and precluded meta-analyses. Results of trials were null for a selection of neurodevelopmental outcomes, however possible benefits of parenteral fish oil supplementation for neurodevelopment was reported in three cohort studies. Certainty of the evidence is hindered by methodological limitations of available trials and observational studies. CONCLUSIONS Further research is required to firmly establish the effects of parenteral fish oil on preterm neurodevelopment.
Collapse
Affiliation(s)
- N Ikeda
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan.
| | - E Shepherd
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - M Makrides
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - A J McPhee
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Neonatal Medicine, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - R A Gibson
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - J F Gould
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; School of Psychology, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
12
|
Gould JF, Roberts RM, Anderson PJ, Makrides M, Sullivan TR, Gibson RA, McPhee AJ, Doyle LW, Bednarz JM, Best KP, Opie G, Travadi J, Cheong JLY, Davis PG, Sharp M, Simmer K, Tan K, Morris S, Lui K, Bolisetty S, Liley H, Stack J, Collins CT. High-Dose Docosahexaenoic Acid in Newborns Born at Less Than 29 Weeks' Gestation and Behavior at Age 5 Years: Follow-Up of a Randomized Clinical Trial. JAMA Pediatr 2024; 178:45-54. [PMID: 37983037 PMCID: PMC10660239 DOI: 10.1001/jamapediatrics.2023.4924] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 11/21/2023]
Abstract
Importance Children born at less than 29 weeks' gestation are at risk of behavioral difficulties. This may be due in part to the lack of transplacental supply of docosahexaenoic acid (DHA), a key fatty acid with structural and functional roles in the brain. Objective To determine whether meeting the neonatal DHA requirement through supplementation is associated with improved behavioral functioning of children born at less than 29 weeks' gestation. Design, Setting and Participants This was a follow-up of children from 10 Australian participating centers in a multi-center, blinded, parallel group randomized clinical trial of infants born at less than 29 weeks' gestation conducted from June 2012 and September 2015, excluding those with additional fatty acid supplementation or major congenital or chromosomal abnormalities. Follow-up took place from August 2018 to May 2021. Parents of surviving children who had not withdrawn from the original trial were invited to complete questionnaires when the child turned 5 years' corrected age. Interventions Infants were randomized to receive daily enteral emulsions providing 60 mg/kg/d of DHA or a soy-oil emulsion (with no DHA) from within the first 3 days of enteral feeding until 36 weeks' postmenstrual age or discharge home, whichever occurred first. Main Outcomes and Measures The primary outcome of this follow-up was parent-rated behavior and emotional functioning as indicated by the Total Difficulties score of the Strengths and Difficulties Questionnaire. Parents also completed questionnaires about their child's behavioral manifestations of executive functioning, as well as a range of health outcomes to assess potential longer-term side effects of DHA intervention. Results Primary outcome data were available for 731 children (76% of 958 surviving eligible children; 361 in the intervention group and 370 in the control group). Of these 731, 452 (47%) were female, and the mean (SD) corrected age at follow-up was 5.4 (0.5) years. Following imputation for missing data, the mean Total Difficulties score was the same in both groups (intervention group, n = 465; mean [SD], 11.8 [6.3]; control group, n = 493; mean [SD], 11.8 [6.0]; mean difference adjusted for sex, gestational age stratum, and hospital, 0.01; 95% CI, -0.87 to 0.89; P = .98). There was no evidence for differences between the groups in any secondary outcomes of behavior, executive functioning, or health. Conclusions and Relevance In this follow-up of a randomized clinical trial, enteral DHA supplementation at the equivalent of the estimated in utero dose for infants born at less than 29 weeks' gestation did not improve behavioral functioning at age 5 years. There were no indications of adverse effects with DHA supplementation. Trial Registration Australian New Zealand Clinical Trial Registry: ACTRN12612000503820.
Collapse
Affiliation(s)
- Jacqueline F. Gould
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, South Australia, Australia
- School of Psychology, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, Adelaide, South Australia, Australia
| | - Rachel M. Roberts
- School of Psychology, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, Adelaide, South Australia, Australia
| | - Peter J. Anderson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Maria Makrides
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, South Australia, Australia
| | - Thomas R. Sullivan
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- School of Public Health, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, South Australia, Australia
| | - Robert A. Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- School of Agriculture, Food and Wine, The University of Adelaide, Waite Campus, Glen Osmond, South Australia, Australia
| | - Andrew J. McPhee
- Neonatal Medicine, Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
| | - Lex W. Doyle
- Department of Obstetrics and Gynaecology, The Royal Women’s Hospital, Parkville, Melbourne, Victoria, Australia
| | - Jana M. Bednarz
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
| | - Karen P. Best
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, South Australia, Australia
| | - Gillian Opie
- Neonatal Services, Mercy Hospital for Women, Heidelberg, Melbourne, Victoria, Australia
| | - Javeed Travadi
- Newborn Services, John Hunter Children’s Hospital, New Lambton Heights, New South Wales, Australia
- Neonatal Intensive Care Unit, Department of Paediatrics, Te Whatu Ora Waikato, Waikato Hospital, Hamilton, New Zealand
| | - Jeanie L. Y. Cheong
- Neonatal Medicine, The Royal Women’s Hospital, Parkville, Melbourne, Victoria, Australia
| | - Peter G. Davis
- Neonatal Medicine, The Royal Women’s Hospital, Parkville, Melbourne, Victoria, Australia
| | - Mary Sharp
- King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - Karen Simmer
- The University of Western Australia, Perth, Western Australia, Australia
| | - Kenneth Tan
- Department of Paediatrics, Monash University, Monash Children’s Hospital, Clayton, Victoria, Australia
| | - Scott Morris
- Neonatal-Perinatal Medicine, Flinders Medical Centre, Flinders Drive, Bedford Park, South Australia, Australia
| | - Kei Lui
- School of Clinical Medicine, Discipline of Paediatrics and Child Health, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Helen Liley
- Mater Research – The Faculty of Medicine, The University of Queensland, South Brisbane, Queensland, Australia
| | - Jacqueline Stack
- Neonatal Intensive Care Unit, Liverpool Hospital, Elizabeth, Liverpool, New South Wales, Australia
| | - Carmel T. Collins
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, South Australia, Australia
| |
Collapse
|
13
|
Wakabayashi N, Haruna M, Yonezawa K, Sasagawa E, Usui Y, Ohori R, Aoyama S, Sasaki S, Nagamatsu T. Association of serum docosahexaenoic acid and eicosapentaenoic acid levels with dietary intakes and supplement use during pregnancy: a prospective observational study. J Nutr Sci 2023; 12:e125. [PMID: 38155803 PMCID: PMC10753464 DOI: 10.1017/jns.2023.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/29/2023] [Accepted: 10/27/2023] [Indexed: 12/30/2023] Open
Abstract
This study aimed to determine the association of serum docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) levels with dietary intakes and supplement use during pregnancy. This prospective observational study was conducted at a university hospital in Tokyo, Japan. Participants in their second and third trimesters were given a self-administered questionnaire assessing the frequency of DHA and EPA supplement use in the past month and a brief-type self-administered diet history questionnaire. Non-fasting serum DHA and EPA levels were analysed using gas chromatography. Differences in biomarkers by frequency of supplement use were determined using multiple comparison analyses, and Spearman's correlation coefficient was used to determine biomarkers and DHA and EPA intakes by food group. Of the 116 participants, 11 (9⋅5 %) in the second trimester and 18 (15⋅5 %) in the third trimester regularly used supplements (≥5 times per week). Regular users had higher serum DHA and EPA levels than never users in the second and third trimesters. Dietary DHA and EPA intake from fish and shellfish was positively correlated with serum DHA and EPA in the second and third trimesters. Supplement use ≥5 times per week and fish and shellfish intake were associated with high serum DHA and EPA levels.
Collapse
Affiliation(s)
- Nana Wakabayashi
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Megumi Haruna
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaori Yonezawa
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Emi Sasagawa
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuriko Usui
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Riko Ohori
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoko Aoyama
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Sasaki
- Department of Social and Preventive Epidemiology, School of Public Health Division of Health Sciences and Nursing, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Facility of Medicine, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
14
|
Gould JF, Bednarz JM, Sullivan TR, McPhee AJ, Gibson RA, Makrides M. Subgroup analyses of a randomized trial of DHA supplementation for infants born preterm with assessments of cognitive development up to 7-years of age: What happens in infants born <29 weeks' gestation? Prostaglandins Leukot Essent Fatty Acids 2023; 198-199:102593. [PMID: 37979339 DOI: 10.1016/j.plefa.2023.102593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/20/2023]
Abstract
A recent trial showed that high-dose docosahexaenoic acid (high-DHA) supplementation of infants born <29 weeks' gestation improves intelligence quotient (IQ) at five years' corrected age. However, this finding has not been detected by other trials of DHA, which either did not measure IQ or included more mature infants. We analyzed the subgroup of 204 infants born <29 weeks' from our earlier randomized trial of high-DHA (∼1 % total fatty acids) or standard-DHA (∼ 0.3 % total fatty acids). Participants were assessed for cognition at 18 months, and IQ and behavior at seven years' corrected age. No group differences were detected for mean cognitive, IQ or behavior scores. At 18 months, 18.8 % of children in the high-DHA group had a cognitive score <85, compared with 31.1 % of children in the standard-DHA group, but at seven years there was no difference. Although an underpowered post-hoc subgroup analysis, this study provides limited support to recommendations that infants born <29 weeks' gestation require supplemental DHA.
Collapse
Affiliation(s)
- J F Gould
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, 72 King William Road, North Adelaide 5006, Australia.
| | - J M Bednarz
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, 72 King William Road, North Adelaide 5006, Australia; School of Psychology & Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, Australia
| | - T R Sullivan
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, 72 King William Road, North Adelaide 5006, Australia; School of Public Health, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide, Australia
| | - A J McPhee
- Neonatal Medicine, Women's and Children's Hospital, 72 King William Road, North Adelaide, Australia
| | - R A Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, 72 King William Road, North Adelaide 5006, Australia; School of Agriculture, Food and Wine, The University of Adelaide, Waite Campus, Glen Osmond, Australia
| | - M Makrides
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, 72 King William Road, North Adelaide 5006, Australia; Discipline of Paediatrics, Faculty of Health and Medical Sciences, The University of Adelaide, North Terrace Adelaide 5000, Australia
| |
Collapse
|
15
|
da Silva ACR, Yadegari A, Tzaneva V, Vasanthan T, Laketic K, Shearer J, Bainbridge SA, Harris C, Adamo KB. Metabolomics to Understand Alterations Induced by Physical Activity during Pregnancy. Metabolites 2023; 13:1178. [PMID: 38132860 PMCID: PMC10745110 DOI: 10.3390/metabo13121178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Physical activity (PA) and exercise have been associated with a reduced risk of cancer, obesity, and diabetes. In the context of pregnancy, maintaining an active lifestyle has been shown to decrease gestational weight gain (GWG) and lower the risk of gestational diabetes mellitus (GDM), hypertension, and macrosomia in offspring. The main pathways activated by PA include BCAAs, lipids, and bile acid metabolism, thereby improving insulin resistance in pregnant individuals. Despite these known benefits, the underlying metabolites and biological mechanisms affected by PA remain poorly understood, highlighting the need for further investigation. Metabolomics, a comprehensive study of metabolite classes, offers valuable insights into the widespread metabolic changes induced by PA. This narrative review focuses on PA metabolomics research using different analytical platforms to analyze pregnant individuals. Existing studies support the hypothesis that exercise behaviour can influence the metabolism of different populations, including pregnant individuals and their offspring. While PA has shown considerable promise in maintaining metabolic health in non-pregnant populations, our comprehension of metabolic changes in the context of a healthy pregnancy remains limited. As a result, further investigation is necessary to clarify the metabolic impact of PA within this unique group, often excluded from physiological research.
Collapse
Affiliation(s)
- Ana Carolina Rosa da Silva
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (A.C.R.d.S.)
| | - Anahita Yadegari
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (A.C.R.d.S.)
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (A.C.R.d.S.)
| | - Tarushika Vasanthan
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5G 2A7, Canada
| | - Katarina Laketic
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Faculty of Kinesiology, Cumming School of Medicine and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Shannon A. Bainbridge
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, Ottawa, ON K1N 6N5, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Cory Harris
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Kristi B. Adamo
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (A.C.R.d.S.)
| |
Collapse
|
16
|
Ali O, Szabó A. Review of Eukaryote Cellular Membrane Lipid Composition, with Special Attention to the Fatty Acids. Int J Mol Sci 2023; 24:15693. [PMID: 37958678 PMCID: PMC10649022 DOI: 10.3390/ijms242115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Biological membranes, primarily composed of lipids, envelop each living cell. The intricate composition and organization of membrane lipids, including the variety of fatty acids they encompass, serve a dynamic role in sustaining cellular structural integrity and functionality. Typically, modifications in lipid composition coincide with consequential alterations in universally significant signaling pathways. Exploring the various fatty acids, which serve as the foundational building blocks of membrane lipids, provides crucial insights into the underlying mechanisms governing a myriad of cellular processes, such as membrane fluidity, protein trafficking, signal transduction, intercellular communication, and the etiology of certain metabolic disorders. Furthermore, comprehending how alterations in the lipid composition, especially concerning the fatty acid profile, either contribute to or prevent the onset of pathological conditions stands as a compelling area of research. Hence, this review aims to meticulously introduce the intricacies of membrane lipids and their constituent fatty acids in a healthy organism, thereby illuminating their remarkable diversity and profound influence on cellular function. Furthermore, this review aspires to highlight some potential therapeutic targets for various pathological conditions that may be ameliorated through dietary fatty acid supplements. The initial section of this review expounds on the eukaryotic biomembranes and their complex lipids. Subsequent sections provide insights into the synthesis, membrane incorporation, and distribution of fatty acids across various fractions of membrane lipids. The last section highlights the functional significance of membrane-associated fatty acids and their innate capacity to shape the various cellular physiological responses.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
| | - András Szabó
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
- HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary
| |
Collapse
|
17
|
Nagel E, Elgersma KM, Gallagher TT, Johnson KE, Demerath E, Gale CA. Importance of human milk for infants in the clinical setting: Updates and mechanistic links. Nutr Clin Pract 2023; 38 Suppl 2:S39-S55. [PMID: 37721461 PMCID: PMC10513735 DOI: 10.1002/ncp.11037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/15/2023] [Accepted: 06/10/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Human milk (HM) is the optimal source of nutrition for infants and has been implicated in multiple aspects of infant health. Although much of the existing literature has focused on the individual components that drive its nutrition content, examining HM as a biological system is needed for meaningful advancement of the field. Investigation of the nonnutritive bioactive components of HM and the maternal, infant, and environmental factors which affect these bioactives is important to better understand the importance of HM provision to infants. This information may inform care of clinical populations or infants who are critically ill, hospitalized, or who have chronic diseases and may benefit most from receiving HM. METHODS In this narrative review, we reviewed literature examining maternal and infant influences on HM composition with a focus on studies published in the last 10 years that were applicable to clinical populations. RESULTS We found multiple studies examining HM components implicated in infant immune and gut health and neurodevelopment. Additional work is needed to understand how donor milk and formula may be used in situations of inadequate maternal HM. Furthermore, a better understanding of how maternal factors such as maternal genetics and metabolic health influence milk composition is needed. CONCLUSION In this review, we affirm the importance of HM for all infants, especially clinical populations. An understanding of how HM composition is modulated by maternal and environmental factors is important to progress the field forward with respect to mechanistic links between HM biology and infant health outcomes.
Collapse
Affiliation(s)
- Emily Nagel
- School of Public Health, University of Minnesota-Twin Cities, Minnesota, USA
| | | | | | - Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minnesota, USA
| | - Ellen Demerath
- School of Public Health, University of Minnesota-Twin Cities, Minnesota, USA
| | - Cheryl A. Gale
- Department of Pediatrics, University of Minnesota-Twin Cities, Minnesota, USA
| |
Collapse
|
18
|
Jiang S, Wang X, Cao T, Kang R, Huang L. Insights on therapeutic potential of clemastine in neurological disorders. Front Mol Neurosci 2023; 16:1279985. [PMID: 37840769 PMCID: PMC10568021 DOI: 10.3389/fnmol.2023.1279985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Clemastine, a Food and Drug Administration (FDA)-approved compound, is recognized as a first-generation, widely available antihistamine that reduces histamine-induced symptoms. Evidence has confirmed that clemastine can transport across the blood-brain barrier and act on specific neurons and neuroglia to exert its protective effect. In this review, we summarize the beneficial effects of clemastine in various central nervous system (CNS) disorders, including neurodegenerative disease, neurodevelopmental deficits, brain injury, and psychiatric disorders. Additionally, we highlight key cellular links between clemastine and different CNS cells, in particular in oligodendrocyte progenitor cells (OPCs), oligodendrocytes (OLs), microglia, and neurons.
Collapse
Affiliation(s)
- Sufang Jiang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xueji Wang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tianyu Cao
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rongtian Kang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lining Huang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Ministry of Education, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Rodrigues RBA, Zafalon RVA, Rentas MF, Risolia LW, Macedo HT, Perini MP, da Silva AMG, Marchi PH, Balieiro JCDC, Mendes WS, Vendramini THA, Brunetto MA. The Supplementation of Docosahexaenoic Acid-Concentrated Fish Oil Enhances Cognitive Function in Puppies. Animals (Basel) 2023; 13:2938. [PMID: 37760338 PMCID: PMC10525578 DOI: 10.3390/ani13182938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Docosahexaenoic acid (DHA) has an important role in brain development and functionality. Therefore, this study aimed to evaluate the effects of DHA-concentrated fish oil on the cognitive function of puppies. Twelve 3-month-old puppies were included, blocked by breed and randomly distributed into two groups: the Control Group (CG), without supplementation, and the Experimental Group (EG), supplemented with 40 mg DHA/kg BW/day. The object discrimination test was used, with a normal stage (NS) and a reverse stage (RS), and blood samples were collected to evaluate the serum polyunsaturated fatty acid (PUFA) concentration and total antioxidant capacity (TAC) before (T0) and 30 (T1), 60 (T2) and 90 (T3) days after beginning the study. For the NS, there were effects of treatment (p = 0.0039) and time (p < 0.0001), in which the correct answer frequency in the EG was higher than the CG. The serum eicosapentaenoic acid (EPA) + HA concentrations at T1, T2 and T3 were higher than at T0 for the EG (p = 0.0159), in addition, EG showed higher serum EPA + DHA concentrations than CG at T2 (p = 0.0245). The TAC values were similar between the groups (p = 0.3211). It was concluded that the cognitive function of puppies can be enhanced with DHA-concentrated fish oil supplementation without increasing the serum lipid oxidation.
Collapse
Affiliation(s)
- Roberta Bueno Ayres Rodrigues
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Rafael Vessecchi Amorim Zafalon
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Mariana Fragoso Rentas
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Larissa Wünsche Risolia
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Henrique Tobaro Macedo
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Mariana Pamplona Perini
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Amanda Maria Gomes da Silva
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Pedro Henrique Marchi
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Júlio César de Carvalho Balieiro
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | | | - Thiago Henrique Annibale Vendramini
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| | - Marcio Antonio Brunetto
- Pet Nutrology Research Center (CEPEN-PET), Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of Sao Paulo (USP), Pirassununga 13635-000, Brazil; (R.B.A.R.); (R.V.A.Z.); (M.F.R.); (L.W.R.); (H.T.M.); (M.P.P.); (A.M.G.d.S.); (P.H.M.); (J.C.d.C.B.)
| |
Collapse
|
20
|
Sherzai D, Moness R, Sherzai S, Sherzai A. A Systematic Review of Omega-3 Fatty Acid Consumption and Cognitive Outcomes in Neurodevelopment. Am J Lifestyle Med 2023; 17:649-685. [PMID: 37711355 PMCID: PMC10498982 DOI: 10.1177/15598276221116052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
INTRODUCTION This systematic review addresses the effects of n-3 long-chain polyunsaturated fatty acids consumption on human neurodevelopment. It evaluates articles published between 2000 and 2022 investigating the cognitive outcomes during the period of neurodevelopment: from fetal development to adolescence. For the purpose of this review the terms LC PUFA and omega-3 fatty acid will be used interchangeably. METHOD Data were sourced from several major databases including PubMed (MEDLINE), Web of Science, and ProQuest Central. Randomized controlled trials (RCTs), nonrandomized controlled trials, prospective or retrospective cohort studies, and observational studies investigating the effects of omega-3 fatty acid consumption from dietary supplements, multiple-nutrient supplement, or food questionnaire on neurodevelopment were considered. Study population was separated in three developmental phases: (1) in-utero, (2) lactation/infancy, and (3) childhood/adolescence. Each article was evaluated for several key factors such as study type, type/dosage of PUFAs, number of subjects, length of intervention, participant age range, population characteristics, outcome measure (both primary/cognitive and secondary/other), results, conclusion, and confounding variables/limitations. RESULTS A total of 88 articles were included in the review, 69 RCTs and 19 longitudinal or observational studies. The results indicate equivocal effect of intervention, with some short-term benefits observed in the areas of visual attention, working memory, executive function, and communication. Omega-3 supplement might have a short-term positive impact on neurodevelopment in all three phases. Supplementation is recommended throughout life, rather than only during the earliest developmental stage.
Collapse
Affiliation(s)
- Dean Sherzai
- Dept of Neurology, Brain Health and Alzheimer's Prevention Program, Loma Linda University, Loma Linda, CA, USA (AS, DS); Oak Ridge High School, El Dorado Hills, CA, USA (RM); California State University, Los Angeles, CA, USA (SS)
| | - Roman Moness
- Dept of Neurology, Brain Health and Alzheimer's Prevention Program, Loma Linda University, Loma Linda, CA, USA (AS, DS); Oak Ridge High School, El Dorado Hills, CA, USA (RM); California State University, Los Angeles, CA, USA (SS)
| | - Sophia Sherzai
- Dept of Neurology, Brain Health and Alzheimer's Prevention Program, Loma Linda University, Loma Linda, CA, USA (AS, DS); Oak Ridge High School, El Dorado Hills, CA, USA (RM); California State University, Los Angeles, CA, USA (SS)
| | - Ayesha Sherzai
- Dept of Neurology, Brain Health and Alzheimer's Prevention Program, Loma Linda University, Loma Linda, CA, USA (AS, DS); Oak Ridge High School, El Dorado Hills, CA, USA (RM); California State University, Los Angeles, CA, USA (SS)
| |
Collapse
|
21
|
Lundgren P, Jacobson L, Gränse L, Hård AL, Sävman K, Hansen-Pupp I, Ley D, Nilsson AK, Pivodic A, Smith LE, Hellström A. Visual outcome at 2.5 years of age in ω-3 and ω-6 long-chain polyunsaturated fatty acid supplemented preterm infants: a follow-up of a randomized controlled trial. THE LANCET REGIONAL HEALTH. EUROPE 2023; 32:100696. [PMID: 37671123 PMCID: PMC10477038 DOI: 10.1016/j.lanepe.2023.100696] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 09/07/2023]
Abstract
Background We investigated ophthalmological outcomes at 2.5 years of corrected age in children born extremely preterm (EPT) to evaluate the effects of postnatal enteral supplementation with ω-3 and ω-6 long-chain polyunsaturated fatty acids. Methods In the Mega Donna Mega clinical trial, EPT infants born at less than 28 weeks of gestation were randomized to receive an enteral supplementation of docosahexaenoic acid (DHA) and arachidonic acid (AA) from birth to 40 weeks postmenstrual age. In this exploratory follow-up at 2.5 years of corrected age, we assessed visual acuity (VA), refraction, manifest strabismus, and nystagmus. Satisfactory VA was defined as ≥20/63. Multiple imputation (MI) was used to address the issue of missing data. Findings Of 178 children in the trial, 115 (with median gestational age (GA) of 25 + 4/7 weeks and median birth weights of 790 g) were ophthalmologically assessed at a median corrected age of 2.7 years (range 2.0-3.9 years). VA assessment was missing in 42.1% (75/178), in 41.7% (35/84) of the AA/DHA supplemented infants, and in 42.6% (40/94) of the control infants. After MI and adjustments for GA, study center, plurality, and corrected age at VA exam, no significant effect of AA/DHA supplementation was detected in VA outcome (≥20/63) (odds ratio 2.16, confidence interval 95% 0.99-4.69, p = 0.053). Interpretation In this randomized controlled trial follow-up, postnatal supplementation with enteral AA/DHA to EPT children did not significantly alter VA at 2.5 years of corrected age. Due to the high loss to follow-up rate and the limited statistical power, additional studies are needed. Funding The Swedish Medical Research Council #2020-01092, The Gothenburg Medical Society, Government grants under the ALF agreement ALFGBG-717971 and ALFGBG-971188, De Blindas Vänner, Knut and Alice Wallenberg Foundation - Wallenberg Clinical Scholars, NIHEY017017, EY030904BCHIDDRC (1U54HD090255 Massachusetts Lions Eye Foundation) supported the study.
Collapse
Affiliation(s)
- Pia Lundgren
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Jacobson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Lotta Gränse
- Department of Clinical Sciences, Ophthalmology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Anna-Lena Hård
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Hansen-Pupp
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anders K Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Aldina Pivodic
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lois E Smith
- The Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
da Costa Silva Kindelan S, Queiroz MP, Barbosa MQ, Viera VB, Guerra GC, Fernandes de Souza Araújo D, Jacielly dos Santos J, Lucia de Azevedo Oliveira M, Milhomens Ferreira Melo PC, Rufino Freitas JC, Gomes Dutra LM, Frazão Tavares de Melo MF, Barbosa Soares JK. Maternal rat prenatal and neonatal treatment with pequi pulp reduces anxiety and lipid peroxidation in brain tissue of rat offspring at adolescence. Heliyon 2023; 9:e19757. [PMID: 37809698 PMCID: PMC10559064 DOI: 10.1016/j.heliyon.2023.e19757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/21/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
The Pequi fruit (Caryocar Brasiliense cambess), typical of the Brazilian cerrado or savannah, is a source of essential fatty acids, carotenoids, and phenolic compounds. The aim of this study was to analyze the effects of consuming this fruit on anxiety behavior and lipid peroxidation in the brains of rats whose mothers were treated (by gavage) during pregnancy and lactation with Pequi fruit (pulp or nuts) at 2000 mg/kg of body weight. Anxiety parameters were assessed using the open field (OF), elevated plus maze (EPM), and light/dark box (LDB) tests. The brain was removed to measure malondialdehyde (MDA) levels. Data were analyzed using One-way Anova (p < 0.05). In the OF, the animals in the pulp group presented more time spent in the central area (20.37 ± 0.73 vs Control: 12.51 ± 0.39; Nuts: 8.28 ± 0.40) and increased locomotion (159.7 ± 6.10) compared to the other groups (Control: 127.3 ± 5.54; Nuts: 139.08 ± 6.57). In the EPM, the pulp group entered into the open arms (8.57 ± 0.36) and stayed more time in the central area (19.44 ± 1.17) compared to the Nuts group (7.14 ± 0.34; 13.00 ± 1.57). In the LDB the pulp group entered more (8.00 ± 0.42 vs Control: 7.16 ± 0.16 and Nuts: 7.42 ± 0.75) and stayed longer in the clear light side (92.18 ± 6.42) than all the other groups (Control: 71.44 ± 3.53; Nuts: 80.57 ± 6.50), respectively. Pulp group presented lower MDA in the brain (55.34 ± 3.04) compared to Control (72.06 ± 4.66) and Nuts (66.57 ± 2.45). We conclude that Pequi pulp consumption during pregnancy and lactation reduces lipid peroxidation in brain tissue and induces anxiolytic-like behavior in rat offspring. These effects were not observed in the Pequi nuts group.
Collapse
Affiliation(s)
- Suedna da Costa Silva Kindelan
- Program of Natural Sciences and Biotechnology, Federal University of Campina Grande, Cuité, Paraiba, Brazil
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | - Michelly Pires Queiroz
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | - Mayara Queiroga Barbosa
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | - Vanessa Bordin Viera
- Program of Natural Sciences and Biotechnology, Federal University of Campina Grande, Cuité, Paraiba, Brazil
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | - Gerlane Coelho Guerra
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | - Jany Jacielly dos Santos
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | | | | | - Juliano Carlo Rufino Freitas
- Program of Natural Sciences and Biotechnology, Federal University of Campina Grande, Cuité, Paraiba, Brazil
- Education and Health Center, Academic Unit of Biology and Chemistry, Federal University of Campina Grande, Cuité, Pariba, Brazil
| | - Larissa Maria Gomes Dutra
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| | | | - Juliana Kessia Barbosa Soares
- Program of Natural Sciences and Biotechnology, Federal University of Campina Grande, Cuité, Paraiba, Brazil
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, Paraiba, Brazil
| |
Collapse
|
23
|
Sullivan TR, Gould JF, Bednarz JM, McPhee AJ, Gibson R, Anderson PJ, Best KP, Sharp M, Cheong JL, Opie GF, Travadi J, Davis PG, Simmer K, Collins CT, Doyle LW, Makrides M. Mediation Analysis to Untangle Opposing Associations of High-Dose Docosahexaenoic Acid With IQ and Bronchopulmonary Dysplasia in Children Born Preterm. JAMA Netw Open 2023; 6:e2317870. [PMID: 37294565 PMCID: PMC10257101 DOI: 10.1001/jamanetworkopen.2023.17870] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Importance High-dose omega-3 docosahexaenoic acid (DHA) supplementation of children born at less than 29 weeks' gestation has been shown to improve IQ despite increasing the risk of bronchopulmonary dysplasia (BPD). Given that BPD is associated with poorer cognitive outcomes, it is unclear whether the increased risk of BPD with DHA supplementation is associated with decreased benefit to IQ. Objective To investigate whether the increased risk of BPD with DHA supplementation was associated with diminished IQ benefit. Design, Setting, and Participants This cohort study used data collected from a multicenter, blinded, randomized controlled trial of DHA supplementation in children born at less than 29 weeks' gestation. Participants were recruited from 2012 to 2015 and followed up until 5 years' corrected age. Data were analyzed from November 2022 to February 2023. Interventions Enteral DHA emulsion (60 mg/kg/d, to match the estimated in-utero requirement) or a control emulsion from the first 3 days of enteral feeds until 36 weeks' postmenstrual age or discharge home. Main Outcomes and Measures Physiological BPD was assessed at 36 weeks' postmenstrual age. IQ was assessed at 5 years' corrected age using the Wechsler Preschool and Primary Scale of Intelligence, 4th Edition; children from the 5 highest-recruiting Australian hospitals were assessed. The total effect of DHA supplementation on IQ was divided into direct and indirect effects using mediation analysis, with BPD as the presumed mediating variable. Results Among 656 surviving children from hospitals involved in IQ follow-up (mean [SD] gestational age at birth, 26.8 [1.4] weeks; 346 males [52.7%]), there were 323 children with DHA supplementation and 333 children in the control group. Mean IQ was 3.45 points (95% CI, 0.38 to 6.53 points) higher in the DHA group than the control group, despite an increase in the risk of BPD (160 children [49.7%] vs 143 children [42.8%] with BPD). The indirect effect of DHA on IQ via BPD was not statistically significant (-0.17 points; 95% CI, -0.62 to 0.13 points), with most of the effect of DHA on IQ occurring independently of BPD (direct effect = 3.62 points; 95% CI, 0.55 to 6.81 points). Conclusions and Relevance This study found that associations of DHA with BPD and IQ were largely independent. This finding suggests that if clinicians supplement children born preterm with high-dose DHA, any resulting increase in BPD risk would not be associated with meaningful reductions in the IQ benefit.
Collapse
Affiliation(s)
- Thomas R. Sullivan
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jacqueline F. Gould
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Psychology, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jana M. Bednarz
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrew J. McPhee
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Neonatal Services, Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
| | - Robert Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Agriculture, Food and Wine, Waite Campus, University of Adelaide, Adelaide, South Australia, Australia
| | - Peter J. Anderson
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Karen P. Best
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Mary Sharp
- King Edward Memorial Hospital, Subiaco, Western Australia, Australia
- Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, Western Australia, Australia
| | - Jeanie L.Y. Cheong
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Newborn Research, Royal Women's Hospital, Parkville, Victoria, Australia
- Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Gillian F. Opie
- Neonatal Services, Mercy Hospital for Women, Melbourne, Victoria, Australia
| | - Javeed Travadi
- Department of Child Health, Neonatal Intensive Care Unit, Waikato Hospital, Waikato, Hamilton, New Zealand
- Newborn Services, John Hunter Children’s Hospital, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter G. Davis
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Newborn Research, Royal Women's Hospital, Parkville, Victoria, Australia
- Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Karen Simmer
- Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, Western Australia, Australia
| | - Carmel T. Collins
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Lex W. Doyle
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Newborn Research, Royal Women's Hospital, Parkville, Victoria, Australia
- Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Maria Makrides
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Discipline of Paediatrics, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Sjöbom U, Andersson MX, Pivodic A, Lund AM, Vanpee M, Hansen-Pupp I, Ley D, Wackernagel D, Sävman K, Smith LEH, Löfqvist C, Hellström A, Nilsson AK. Modification of serum fatty acids in preterm infants by parenteral lipids and enteral docosahexaenoic acid/arachidonic acid: A secondary analysis of the Mega Donna Mega trial. Clin Nutr 2023; 42:962-971. [PMID: 37120902 PMCID: PMC10512593 DOI: 10.1016/j.clnu.2023.04.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND & AIM Preterm infants risk deficits of long-chain polyunsaturated fatty acids (LCPUFAs) that may contribute to morbidities and hamper neurodevelopment. We aimed to determine longitudinal serum fatty acid profiles in preterm infants and how the profiles are affected by enteral and parenteral lipid sources. METHODS Cohort study analyzing fatty acid data from the Mega Donna Mega study, a randomized control trial with infants born <28 weeks of gestation (n = 204) receiving standard nutrition or daily enteral lipid supplementation with arachidonic acid (AA):docosahexaenoic acid (DHA) (100:50 mg/kg/day). Infants received an intravenous lipid emulsion containing olive oil:soybean oil (4:1). Infants were followed from birth to postmenstrual age 40 weeks. Levels of 31 different fatty acids from serum phospholipids were determined by GC-MS and reported in relative (mol%) and absolute concentration (μmol l-1) units. RESULTS Higher parenteral lipid administration resulted in lower serum proportion of AA and DHA relative to other fatty acids during the first 13 weeks of life (p < 0.001 for the 25th vs the 75th percentile). The enteral AA:DHA supplement increased the target fatty acids with little impact on other fatty acids. The absolute concentration of total phospholipid fatty acids changed rapidly in the first weeks of life, peaking at day 3, median (Q1-Q3) 4452 (3645-5466) μmol l-1, and was positively correlated to the intake of parenteral lipids. Overall, infants displayed common fatty acid trajectories over the study period. However, remarkable differences in fatty acid patterns were observed depending on whether levels were expressed in relative or absolute units. For example, the relative levels of many LCPUFAs, including DHA and AA, declined rapidly after birth while their absolute concentrations increased in the first week of life. For DHA, absolute levels were significantly higher compared to cord blood from day 1 until postnatal week 16 (p < 0.001). For AA, absolute postnatal levels were lower compared to cord blood from week 4 throughout the study period (p < 0.05). CONCLUSIONS Our data show that parenteral lipids aggravate the postnatal loss of LCPUFAs seen in preterm infants and that serum AA available for accretion is below that in utero. Further research is needed to establish optimal postnatal fatty acid supplementation and profiles in extremely preterm infants to promote development and long-term health. CLINICAL TRIAL REGISTRY ClinicalTrials.gov, identifier: NCT03201588.
Collapse
Affiliation(s)
- Ulrika Sjöbom
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Learning and Leadership for Health Care Professionals at the Institute of Health and Care Science at Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Mats X Andersson
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden.
| | - Aldina Pivodic
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Anna-My Lund
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden.
| | - Mireille Vanpee
- Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| | - Ingrid Hansen-Pupp
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden.
| | - David Ley
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden.
| | - Dirk Wackernagel
- Department of Neonatology, Karolinska University Hospital and Institute, Astrid Lindgrens Children's Hospital, Stockholm, Sweden.
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Department of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Lois E H Smith
- The Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Chatarina Löfqvist
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Learning and Leadership for Health Care Professionals at the Institute of Health and Care Science at Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Anders K Nilsson
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
25
|
Wu CH, Hsu WL, Tsai CC, Chao HR, Wu CY, Chen YH, Lai YR, Chen CH, Tsai MH. 7,10,13,16-Docosatetraenoic acid impairs neurobehavioral development by increasing reactive oxidative species production in Caenorhabditis elegans. Life Sci 2023; 319:121500. [PMID: 36796717 DOI: 10.1016/j.lfs.2023.121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
AIMS To investigate human breast milk (HBM) lipids that may adversely affect infant neurodevelopment. MAIN METHODS We performed multivariate analyses that combined lipidomics and psychologic Bayley-III scales to identify which HBM lipids are involved in regulating infant neurodevelopment. We observed a significant moderate negative correlation between 7,10,13,16-docosatetraenoic acid (omega-6, C22H36O2, the common name adrenic acid, AdA) and adaptive behavioral development. We further studied the effects of AdA on neurodevelopment by using Caenorhabditis elegans (C. elegans) as a model. Worms from larval stages L1 to L4 were supplemented with AdA at 5 nominal concentrations (0 μM [control], 0.1 μM, 1 μM, 10 μM, and 100 μM) and subjected to behavioral and mechanistic analyses. KEY FINDINGS Supplementation with AdA from larval stages L1 to L4 impaired neurobehavioral development, such as locomotive behaviors, foraging ability, chemotaxis behavior, and aggregation behavior. Furthermore, AdA upregulated the production of intracellular reactive oxygen species. AdA-induced oxidative stress blocked serotonin synthesis and serotoninergic neuron activity and inhibited expression of daf-16 and the daf-16-regulated genes mtl-1, mtl-2, sod-1, and sod-3, resulting in attenuation of the lifespan in C. elegans. SIGNIFICANCE Our study reveals that AdA is a harmful HBM lipid that may have adverse effects on infant adaptive behavioral development. We believe this information may be critical for AdA administration guidance in children's health care.
Collapse
Affiliation(s)
- Chia-Hsiu Wu
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wen-Li Hsu
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ching-Chung Tsai
- Department of Pediatrics, E-Da Hospital, No. 8, Yida Rd., Kaohsiung 82445, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - How-Ran Chao
- Department of Environmental Science and Engineering, College of Engineering, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Emerging Compounds Research Center, General Research Service Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Institute of Food Safety Management, College of Agriculture, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan.
| | - Ching-Ying Wu
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan; Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan.
| | - Yi-Hsuan Chen
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yun-Ru Lai
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chu-Huang Chen
- Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX 77030, USA; New York Heart Research Foundation, Mineola, NY 11501, USA; Institute for Biomedical Sciences, Shinshu University, Nagano 390-8621, Japan.
| | - Ming-Hsien Tsai
- Department of Child Care, College of Humanities and Social Sciences, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Department of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
26
|
Khoury S, Soubeyre V, Cabaret S, Grégoire S, Mézière E, Masson E, Grosmaitre X, Bretillon L, Berdeaux O, Acar N, Le Bon AM. Impact of dietary n-3 polyunsaturated fatty acid intake during the perinatal and post-weaning periods on the phospholipid and ganglioside composition of olfactory tissues. Prostaglandins Leukot Essent Fatty Acids 2023; 191:102556. [PMID: 36870298 DOI: 10.1016/j.plefa.2023.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/02/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
The olfactory mucosa (OM) and olfactory bulb (OB) are neuronal tissues that contribute to the early processing of olfactory information. They contain significant amounts of n-3 and n-6 polyunsaturated fatty acids (PUFAs), which are crucial for neuronal tissue development. In this study, we evaluated the impact of feeding mice diets that are either deficient in α-linolenic acid (ALA) or supplemented with n-3 long-chain PUFAs from gestation to adolescence on the phospholipid and ganglioside composition of these tissues. Both diets modified the levels of some phospholipid classes, notably the phosphatidylserine and phosphatidylethanolamine levels. In addition, the low-ALA diet enriched n-6 PUFAs in the main phospholipid classes of both tissues, while the diet supplemented with n-3 PUFAs enhanced the n-3 PUFA-containing phospholipid species level, mainly in OM. The diets also modulated the levels and profiles of several ganglioside classes in OM and OB. These modifications may have repercussions on the olfactory sensitivity.
Collapse
Affiliation(s)
- Spiro Khoury
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France; INRAE, PROBE Research Infrastructure, ChemoSens facility, F-21000 Dijon, France
| | - Vanessa Soubeyre
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Stéphanie Cabaret
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France; INRAE, PROBE Research Infrastructure, ChemoSens facility, F-21000 Dijon, France
| | - Stéphane Grégoire
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Esther Mézière
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France; INRAE, PROBE Research Infrastructure, ChemoSens facility, F-21000 Dijon, France
| | - Elodie Masson
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Xavier Grosmaitre
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Lionel Bretillon
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Olivier Berdeaux
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France; INRAE, PROBE Research Infrastructure, ChemoSens facility, F-21000 Dijon, France
| | - Niyazi Acar
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Anne Marie Le Bon
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, F-21000 Dijon, France.
| |
Collapse
|
27
|
K B C, R H, R A, G JJB, J P D, Banjara SK, M A, K PR, B N, M SP. Macronutrient composition of term and preterm human milk of different socio economic groups. Prostaglandins Leukot Essent Fatty Acids 2023; 192:102571. [PMID: 37079956 DOI: 10.1016/j.plefa.2023.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
Assessment of the nutritional composition of Human Breast Milk (HBM) is important to understand its sufficiency as the sole nutrient source in infants. The present study is aimed to analyze the proximate composition along with total amino acid and fatty acid profile in term and preterm HBM of different socio economic status. This cross sectional study included, 120 lactating mothers with term or preterm gestation from maternity hospitals located in Hyderabad, Telangana. Nutritional proximate, total amino and fatty acid profiles were estimated in pooled human milk collected from each participant within the first week of postpartum. The macronutrient composition in term was similar to that of preterm breast milk. The essential amino acid Leucine was significantly high in preterm (8.91 ± 0.18) when compared to term (8.61 ± 0.23). ω-5 fatty acid Myristoleic acid was significantly high in preterm (0.14 ± 0.02) when compared to term (0.11 ± 0.02), whereas ω-6 fatty acids like Docosadienoic Acid and Eicosadienoic acid were found to be significantly high in term when compared to preterm. Further, it was also found that the mono unsaturated and ω-9 fatty acids were significantly high in lower socio economic group, whereas, poly unsaturated and ω -3 and 6 fatty acids were significantly high in upper socio economic group. The present study concludes that, nutritional composition like essential amino and fatty acids of human milk vary significantly between different gestational age as well as in socio economic groups.
Collapse
Affiliation(s)
- Chathyushya K B
- Clinical Epidemiology Division, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Hemalatha R
- Director. ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Ananthan R
- Food Chemistry Division, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - J J Babu G
- Clinical Epidemiology Division, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Devraj J P
- Clinical Epidemiology Division, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Santosh Kumar Banjara
- Maternal and Child Health Division, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Alimelu M
- Department of Neonatology, Niloufer Hospital, Osmania Medical College, Hyderabad, Telangana, India
| | - Pradeep Reddy K
- Neonatology, Rainbow Children Hospital, Banjarahills, Hyderabad, Telangana, India
| | - Nitasha B
- Neonatology, Rainbow Children Hospital, Banjarahills, Hyderabad, Telangana, India
| | - Shiva Prakash M
- Former Scientist, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India.
| |
Collapse
|
28
|
Capra ME, Stanyevic B, Giudice A, Monopoli D, Decarolis NM, Esposito S, Biasucci G. Long-Chain Polyunsaturated Fatty Acids Effects on Cardiovascular Risk in Childhood: A Narrative Review. Nutrients 2023; 15:nu15071661. [PMID: 37049503 PMCID: PMC10096679 DOI: 10.3390/nu15071661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Long-chain polyunsaturated fatty acids (LCPUFAs) are semi-essential fatty acids widely studied in adult subjects for their healthy-heart effects, especially on secondary prevention in patients who already experienced a cardiac event. LCPUFAs consumption is safe, without adverse effects, and they are usually well-tolerated; they can be taken either in foods or as nutritional supplements. LCPUFAs' positive effect on global health has been worldwide recognized also for pediatric patients. In childhood and adolescence, research has mainly focused on LCPUFAs' effects on neurodevelopment, brain and visual functions and on maternal-fetal medicine, yet their cardiovascular effects in childhood are still understudied. Atherosclerosis is a multifactorial process that starts even before birth and progresses throughout life; thus, cardiovascular prevention is advisable and effective from the very first years of life. Nutritional and lifestyle interventions are the main factors that can interfere with atherosclerosis in childhood, and the consumption of specific nutrients, such as LCPUFAs, can enhance positive nutritional effects. The aim of our narrative review is to analyze the effect of LCPUFAs on cardiovascular risk factors and on cardiovascular risk prevention in developmental age, focusing on specific conditions such as weight excess and dyslipidemia.
Collapse
Affiliation(s)
- Maria Elena Capra
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
- Società Italiana di Nutrizione Pediatrica, 20126 Milan, Italy
| | - Brigida Stanyevic
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Antonella Giudice
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Delia Monopoli
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Nicola Mattia Decarolis
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
- Società Italiana di Nutrizione Pediatrica, 20126 Milan, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
29
|
Küster A, Croyal M, Moyon T, Darmaun D, Ouguerram K, Ferchaud-Roucher V. Characterization of lipoproteins and associated lipidome in very preterm infants: a pilot study. Pediatr Res 2023; 93:938-947. [PMID: 35739258 DOI: 10.1038/s41390-022-02159-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Preterm birth is associated with higher risks of suboptimal neurodevelopment and cardiometabolic disease later in life. Altered maternal-fetal lipid supply could play a role in such risks. Our hypothesis was that very preterm infants born with very low birth weight (VLBW) have altered lipidome and apolipoprotein profiles, compared with term infants. METHODS Seven mothers of VLBW infants born at <32 GA and 8 full-term mother-infant dyads were included. Cholesterol and triglycerides in lipoproteins were determined in maternal plasma and in the two blood vessels of the umbilical cord (vein (UV) and artery (UA)) following FPLC isolation. Apolipoprotein concentrations in lipoproteins and plasma lipidomic analysis were performed by LC-MS/MS. RESULTS We found higher cholesterol and VLDL-cholesterol in UV and UA and lower apolipoprotein A-I in HDL2 in UV in preterm neonates. Phosphatidylcholine (PC) containing saturated and monounsaturated fatty acids and specific sphingomyelin species were increased in UV and UA, whereas PC containing docosahexaenoic acid (DHA) was reduced in UV of VLBW neonates. CONCLUSIONS Lower DHA-PC suggests a lower DHA bioavailability and may contribute to the impaired neurodevelopment. Altered HDL-2, VLDL, and sphingomyelin profile reflect an atherogenic risk and increased metabolic risk at adulthood in infants born prematurely. IMPACT Lower ApoA-I in HDL2, and increased specific sphingomyelin and phosphatidylcholine containing saturated and monounsaturated fatty acid could explain the accumulation of cholesterol in umbilical vein in VLBW preterm neonates. Decreased phosphatidylcholine containing DHA suggest a reduced DHA availability for brain development in VLBW preterm infants. Characterization of alterations in fetal lipid plasma and lipoprotein profiles may help to explain at least in part the causes of the elevated cardiovascular risk known in people born prematurely and may suggest that a targeted nutritional strategy based on the composition of fatty acids carried by phosphatidylcholine may be promising in infants born very early.
Collapse
Affiliation(s)
- Alice Küster
- Nantes University INRAe, UMR 1280 PhAN, CHU Nantes, CRNH Ouest, IMAD, 44000, Nantes, France
- Division of Inborn Errors of Metabolism and Neurometabolism, CHU Nantes, INSERM, Centre d'Investigation Clinique, 44000, Nantes, France
| | - Mikael Croyal
- Nantes Université, CNRS, INSERM, l'institut du Thorax, 44000, Nantes, France
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 44000, Nantes, France
- CRNH-Ouest Mass Spectrometry Core Facility, 44000, Nantes, France
| | - Thomas Moyon
- Nantes University INRAe, UMR 1280 PhAN, CHU Nantes, CRNH Ouest, IMAD, 44000, Nantes, France
| | - Dominique Darmaun
- Nantes University INRAe, UMR 1280 PhAN, CHU Nantes, CRNH Ouest, IMAD, 44000, Nantes, France
| | - Khadija Ouguerram
- Nantes University INRAe, UMR 1280 PhAN, CHU Nantes, CRNH Ouest, IMAD, 44000, Nantes, France
| | | |
Collapse
|
30
|
Thanhaeuser M, Steyrl D, Fuiko R, Brandstaetter S, Binder C, Thajer A, Huber-Dangl M, Haiden N, Berger A, Repa A. A secondary Outcome Analysis of a Randomized Trial Using a Mixed Lipid Emulsion Containing Fish Oil in Infants with Extremely Low Birth Weight: Cognitive and Behavioral Outcome at Preschool Age. J Pediatr 2023; 254:68-74.e3. [PMID: 36257349 DOI: 10.1016/j.jpeds.2022.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To evaluate the impact of a parenteral lipid emulsion containing fish oil compared with a soybean oil based-lipid emulsion on the cognitive outcome and behavior of preschool children with extremely low birth weight. STUDY DESIGN This was a retrospective secondary outcome analysis of a randomized controlled trial performed between June 2012 and June 2015. Infants with extremely low birth weight received either a mixed (soybean oil, medium chain triglycerides, olive oil, fish oil) or a soybean oil-based lipid emulsion for parenteral nutrition. Data from the Kaufman Assessment Battery for Children II, the Child Behavior Checklist 1.5-5, and anthropometry were collected from medical charts at 5.6 years of age. RESULTS At discharge, 206 of the 230 study participants were eligible. At 5 years 6 months of age, data of 153 of 206 infants (74%) were available for analysis. There were no significant differences in Kaufman Assessment Battery for Children II scores for Sequential/Gsm, Simultaneous/Gv, Learning/Glr, and Mental Processing Index (mixed lipid: median, 97.5 [IQR, 23.5]; soybean oil: median, 96 [IQR, 19.5]; P = .43) or Child Behavior Checklist 1.5-5 scores for internalizing problems, externalizing problems, or total problems (mixed lipid: median, 37 [IQR, 12.3]; soybean oil: median, 37 [IQR, 13.5]; P = .54). CONCLUSIONS A RandomForest machine learning regression analysis did not show an effect of type of lipid emulsion on cognitive and behavioral outcome. Parenteral nutrition using a mixed lipid emulsion containing fish oil did not affect neurodevelopment and had no impact on child behavior of infants with extremely low birth weights at preschool age. TRIAL REGISTRATION ClinicalTrials.gov: NCT01585935.
Collapse
Affiliation(s)
- Margarita Thanhaeuser
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria.
| | - David Steyrl
- Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria
| | - Renate Fuiko
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Sophia Brandstaetter
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Christoph Binder
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Alexandra Thajer
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Mercedes Huber-Dangl
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Andreas Repa
- Division of Neonatology, Pediatric Intensive Care Medicine and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Rizzo G, Baroni L, Lombardo M. Promising Sources of Plant-Derived Polyunsaturated Fatty Acids: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1683. [PMID: 36767052 PMCID: PMC9914036 DOI: 10.3390/ijerph20031683] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 06/01/2023]
Abstract
(1) Background: Polyunsaturated fatty acids (PUFAs) are known for their ability to protect against numerous metabolic disorders. The consumption of oily fish is the main source of PUFAs in human nutrition and is commonly used for supplement production. However, seafood is an overexploited source that cannot be guaranteed to cover the global demands. Furthermore, it is not consumed by everyone for ecological, economic, ethical, geographical and taste reasons. The growing demand for natural dietary sources of PUFAs suggests that current nutritional sources are insufficient to meet global needs, and less and less will be. Therefore, it is crucial to find sustainable sources that are acceptable to all, meeting the world population's needs. (2) Scope: This review aims to evaluate the recent evidence about alternative plant sources of essential fatty acids, focusing on long-chain omega-3 (n-3) PUFAs. (3) Method: A structured search was performed on the PubMed search engine to select available human data from interventional studies using omega-3 fatty acids of non-animal origin. (4) Results: Several promising sources have emerged from the literature, such as algae, microorganisms, plants rich in stearidonic acid and GM plants. However, the costs, acceptance and adequate formulation deserve further investigation.
Collapse
Affiliation(s)
- Gianluca Rizzo
- Independent Researcher, Via Venezuela 66, 98121 Messina, Italy
| | - Luciana Baroni
- Scientific Society for Vegetarian Nutrition, 30171 Venice, Italy
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, 00166 Rome, Italy
| |
Collapse
|
32
|
Best KP, Sullivan TR, Gunaratne AW, Gould JF, Gibson RA, Collins CT, Makrides M, Green TJ. Effect of Docosahexaenoic Acid (DHA) Supplementation of Preterm Infants on Growth, Body Composition, and Blood Pressure at 7-Years Corrected Age: Follow-Up of a Randomized Controlled Trial. Nutrients 2023; 15:nu15020335. [PMID: 36678206 PMCID: PMC9867194 DOI: 10.3390/nu15020335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Aim: To determine if supplementation of infants born <33 weeks’ gestation with higher dose docosahexaenoic acid (DHA) affects growth, body composition, and blood pressure at 7 y corrected age (CA) and if treatment effects differed by infant sex at birth and birth weight strata (<1250 and ≥1250 g). Methods: Seven-year follow-up of an Australian multicenter randomized controlled trial in which 657 infants were fed high-DHA (≈1% total fatty acids) enteral feeds or standard-DHA (≈0.3% total fatty acids) from age 2−4 d until term CA. Seven-year CA outcomes were growth (weight, height), body composition (lean body mass, fat mass, waist, and hip circumference), and blood pressure. Results: There was no effect of high-DHA enteral feeds compared with standard-DHA on growth, body composition, and blood pressure at 7-year CA either overall or in subgroup analysis by sex. There was a significant interaction between high-DHA and birthweight strata on height at 7-y CA (p = 0.03). However, the post-hoc analyses by birthweight strata did not reach significance (p > 0.1). High-DHA group infants were more likely to be classified as obese (relative risk 1.6 (95% CI 1.0, 2.6); p = 0.05). Conclusions: DHA supplementation of premature infants did not affect growth, body composition, or blood pressure at 7-year CA overall by sex and birthweight strata. The finding of a higher risk of obesity in children who receive high-DHA needs to be interpreted with caution due to the small number of children classified as obese.
Collapse
Affiliation(s)
- Karen P. Best
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Thomas R. Sullivan
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Public Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Anoja W. Gunaratne
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Jacqueline F. Gould
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Robert A. Gibson
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- School of Agriculture Food and Wine, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Carmel T. Collins
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Maria Makrides
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Tim J. Green
- SAHMRI Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
- Correspondence: ; Tel.: +61-8-8161-6179
| |
Collapse
|
33
|
James G, Stephenson K, Callaghan-Gillespie M, Kamara MT, Park HG, Brenna JT, Manary MJ. Docosahexaenoic Acid Stability in Ready-to-Use Therapeutic Food. Foods 2023; 12:foods12020308. [PMID: 36673399 PMCID: PMC9858440 DOI: 10.3390/foods12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Ready-to-use therapeutic food (RUTF) is used to treat young children diagnosed with severe acute malnutrition. RUTF with low and balanced linoleic and alpha-linolenic acid, plus omega-3 docosahexaenoic acid (DHA), supports long-term cognitive recovery. DHA is prone to degradation due to peroxidation, possibly exacerbated by the iron inherently in RUTF. Our goals were to prepare benchtop and manufacturing scale of RUTF formulations that include DHA and measure its retention. Twenty-seven RUTF formulas with base ingredients, including oats, high oleic or commodity peanuts, and encapsulated or oil-based DHA at various levels were prepared at benchtop scale, followed by seven months of climate-controlled storage. These pilot samples had similar relative DHA retention. At the manufacturing scale, DHA was added at one of two stages in the process, either at the initial or the final mixing stage. Samples taken at preliminary or later steps show that less than 20% of DHA added at the early stages disappeared prior to packaging for any recipe tested. Overall, our data indicate that most DHA included in RUTF is retained in the final product and that DHA is best retained when added at the latest manufacturing stage.
Collapse
Affiliation(s)
- Genevieve James
- Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - Kevin Stephenson
- Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | | | | | - Hui Gyu Park
- Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - J. Thomas Brenna
- Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, USA
| | - Mark J. Manary
- Department of Pediatrics, Washington University, St. Louis, MO 63110, USA
- Project Peanut Butter, Freetown 47235, Sierra Leone
- Children’s Nutrition Research Center, USDA-Agricultural Research Service, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-(314)-454-2178
| |
Collapse
|
34
|
Wang Y, Feltham BA, Louis XL, Eskin MNA, Suh M. Maternal diets affected ceramides and fatty acids in brain regions of neonatal rats with prenatal ethanol exposure. Nutr Neurosci 2023; 26:60-71. [PMID: 34957933 DOI: 10.1080/1028415x.2021.2017661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objectives: Ceramide (Cer), known as apoptotic markers, increases with prenatal ethanol (EtOH) exposure, resulting in neuroapoptosis. Whether maternal nutrition can impact Cer concentrations in brain, via altering plasma and brain fatty acid compositions have not been examined. This study compared a standard chow with a formulated semi-purified energy dense (E-dense) diet on fatty acid composition, Cer concentrations, and apoptosis in plasma and brain regions (cortex, cerebellum, and hippocampus) of pups exposed to EtOH during gestation. Methods: Pregnant Sprague-Dawley rats were randomized into four groups: chow (n = 6), chow + EtOH (20% v/v) (n = 7), E-dense (n = 6), and E-dense + EtOH (n = 8). At postnatal day 7, representing the peak brain growth spurt in rats, lipids, and apoptosis were analyzed by gas chromatography and a fluorometric caspase-3 assay kit, respectively. Results: Maternal E-dense diet increased total fatty acid concentrations (p < 0.0001), including docosahexaenoic acid (DHA) (p < 0.0001) in plasma, whereas DHA concentrations were decreased in the cerebellum (p < 0.03) of pups than those from chow-fed dams. EtOH-induced Cer elevations in the hippocampus of pups born to dams fed chow were reduced by an E-dense diet (p < 0.02). No significant effects of maternal diet quality and EtOH were observed on caspase-3 activity. No significant correlations existed between plasma/brain fatty acids and Cer concentrations. Discussions: Maternal diet quality affected fatty acid compositions and Cer concentrations of pups with prenatal EtOH exposure, differently. Maternal nutrition has the potential to prevent or alleviate some of the adverse effects of prenatal EtOH exposure.
Collapse
Affiliation(s)
- Yidi Wang
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Bradley A Feltham
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Xavier L Louis
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Michael N A Eskin
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Miyoung Suh
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| |
Collapse
|
35
|
Donovan SM, Abrams SA, Azad MB, Belfort MB, Bode L, Carlson SE, Dallas DC, Hettinga K, Järvinen K, Kim JH, Lebrilla CB, McGuire MK, Sela DA, Neu J. Summary of the joint National Institutes of Health and the Food and Drug Administration workshop titled "exploring the science surrounding the safe use of bioactive ingredients in infant formula: Considerations for an assessment framework". J Pediatr 2022; 255:30-41.e1. [PMID: 36463938 PMCID: PMC10121942 DOI: 10.1016/j.jpeds.2022.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/20/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL
| | - Steven A Abrams
- Department of Pediatrics Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada; Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Mandy B Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, Kansas University Medical Center and The University of Kansas, Kansas City, KS
| | - David C Dallas
- Department of Nutrition, Oregon State University, Corvallis, OR
| | - Kasper Hettinga
- Department of Food Sciences and Agrotechnology, Wageningen University, Wageningen, Netherlands
| | - Kirsi Järvinen
- Department of Pediatrics, Golisano Children's Hospital and University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Jae H Kim
- Perinatal Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH
| | | | | | - David A Sela
- Department of Food Science, University of Massachusetts, Amherst, Amherst, MA
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL.
| |
Collapse
|
36
|
Gould JF, Makrides M, Gibson RA, Sullivan TR, McPhee AJ, Anderson PJ, Best KP, Sharp M, Cheong JLY, Opie GF, Travadi J, Bednarz JM, Davis PG, Simmer K, Doyle LW, Collins CT. Neonatal Docosahexaenoic Acid in Preterm Infants and Intelligence at 5 Years. N Engl J Med 2022; 387:1579-1588. [PMID: 36300974 DOI: 10.1056/nejmoa2206868] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) is a component of neural tissue. Because its accretion into the brain is greatest during the final trimester of pregnancy, infants born before 29 weeks' gestation do not receive the normal supply of DHA. The effect of this deficiency on subsequent cognitive development is not well understood. METHODS We assessed general intelligence at 5 years in children who had been enrolled in a trial of neonatal DHA supplementation to prevent bronchopulmonary dysplasia. In the previous trial, infants born before 29 weeks' gestation had been randomly assigned in a 1:1 ratio to receive an enteral emulsion that provided 60 mg of DHA per kilogram of body weight per day or a control emulsion from the first 3 days of enteral feeds until 36 weeks of postmenstrual age or discharge home, whichever occurred first. Children from 5 of the 13 centers in the original trial were invited to undergo assessment with the Wechsler Preschool and Primary Scale of Intelligence (WPPSI) at 5 years of corrected age. The primary outcome was the full-scale intelligence quotient (FSIQ) score. Secondary outcomes included the components of WPPSI. RESULTS A total of 1273 infants underwent randomization in the original trial; of the 656 surviving children who had undergone randomization at the centers included in this follow-up study, 480 (73%) had an FSIQ score available - 241 in the DHA group and 239 in the control group. After imputation of missing data, the mean (±SD) FSIQ scores were 95.4±17.3 in the DHA group and 91.9±19.1 in the control group (adjusted difference, 3.45; 95% confidence interval, 0.38 to 6.53; P = 0.03). The results for secondary outcomes generally did not support that obtained for the primary outcome. Adverse events were similar in the two groups. CONCLUSIONS In infants born before 29 weeks' gestation who had been enrolled in a trial to assess the effect of DHA supplementation on bronchopulmonary dysplasia, the use of an enteral DHA emulsion until 36 weeks of postmenstrual age was associated with modestly higher FSIQ scores at 5 years of age than control feeding. (Funded by the Australian National Health and Medical Research Council and Nu-Mega Ingredients; N3RO Australian New Zealand Clinical Trials Registry number, ACTRN12612000503820.).
Collapse
Affiliation(s)
- Jacqueline F Gould
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Maria Makrides
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Robert A Gibson
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Thomas R Sullivan
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Andrew J McPhee
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Peter J Anderson
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Karen P Best
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Mary Sharp
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Jeanie L Y Cheong
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Gillian F Opie
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Javeed Travadi
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Jana M Bednarz
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Peter G Davis
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Karen Simmer
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Lex W Doyle
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| | - Carmel T Collins
- From SAHMRI Women and Kids, South Australian Health and Medical Research Institute (J.F.G., M.M., R.A.G., T.R.S., A.J.M., K.P.B., J.M.B., C.T.C.), the Schools of Medicine (J.F.G., M.M., A.J.M., K.P.B., C.T.C.), Psychology (J.F.G.), Agriculture, Food, and Wine (R.A.G.), and Public Health (T.R.S.), University of Adelaide, and the Department of Neonatal Medicine, Women's and Children's Hospital (A.J.M.), Adelaide, SA, Newborn Research, Royal Women's Hospital (J.L.Y.C., P.G.D., L.W.D.), the Murdoch Children's Research Institute (P.J.A., J.L.Y.C., P.G.D., L.W.D.), and the Departments of Obstetrics and Gynaecology (J.L.Y.C., P.G.D., L.W.D.) and Paediatrics (L.W.D.), University of Melbourne, the School of Psychological Sciences, Monash University (P.J.A.), and the Department of Paediatrics, Mercy Hospital for Women (G.F.O.), Melbourne, VIC, King Edward Memorial Hospital, Subiaco, WA (M.S.), Newborn Medicine, Centre for Neonatal Research and Education, University of Western Australia, Perth, WA (M.S., K.S.), and the Neonatal Intensive Care Unit, John Hunter Children's Hospital, New Lambton Heights, NSW (J.T.) - all in Australia
| |
Collapse
|
37
|
Altered Cord Blood Lipid Concentrations Correlate with Birth Weight and Doppler Velocimetry of Fetal Vessels in Human Fetal Growth Restriction Pregnancies. Cells 2022; 11:cells11193110. [PMID: 36231072 PMCID: PMC9562243 DOI: 10.3390/cells11193110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Fetal growth restriction (FGR) is associated with short- and long-term morbidity, often with fetal compromise in utero, evidenced by abnormal Doppler velocimetry of fetal vessels. Lipids are vital for growth and development, but metabolism in FGR pregnancy, where fetuses do not grow to full genetic potential, is poorly understood. We hypothesize that triglyceride concentrations are increased in placentas and that important complex lipids are reduced in cord plasma from pregnancies producing the smallest babies (birth weight < 5%) and correlate with ultrasound Dopplers. Dopplers (umbilical artery, UA; middle cerebral artery, MCA) were assessed longitudinally in pregnancies diagnosed with estimated fetal weight (EFW) < 10% at ≥29 weeks gestation. For a subset of enrolled women, placentas and cord blood were collected at delivery, fatty acids were extracted and targeted lipid class analysis (triglyceride, TG; phosphatidylcholine, PC; lysophosphatidylcholine, LPC; eicosanoid) performed by LCMS. For this sub-analysis, participants were categorized as FGR (Fenton birth weight, BW ≤ 5%) or SGA "controls" (Fenton BW > 5%). FGRs (n = 8) delivered 1 week earlier (p = 0.04), were 29% smaller (p = 0.002), and had 133% higher UA pulsatility index (PI, p = 0.02) than SGAs (n = 12). FGR plasma TG, free arachidonic acid (AA), and several eicosanoids were increased (p < 0.05); docosahexaenoic acid (DHA)-LPC was decreased (p < 0.01). Plasma TG correlated inversely with BW (p < 0.05). Plasma EET, non-esterified AA, and DHA correlated inversely with BW and directly with UA PI (p < 0.05). Placental DHA-PC and AA-PC correlated directly with MCA PI (p < 0.05). In fetuses initially referred for inadequate fetal growth (EFW < 10%), those with BW ≤ 5% demonstrated distinctly different cord plasma lipid profiles than those with BW > 5%, which correlated with Doppler PIs. This provides new insights into fetal lipidomic response to the FGR in utero environment. The impact of these changes on specific processes of growth and development (particularly fetal brain) have not been elucidated, but the relationship with Doppler PI may provide additional context for FGR surveillance, and a more targeted approach to nutritional management of these infants.
Collapse
|
38
|
Asfour SS, Alshaikh B, AlMahmoud L, Sumaily HH, Alodhaidan NA, Alkhourmi M, Abahussain HA, Khalil TM, Albeshri BA, Alhamidi AA, Al-Anazi MR, Asfour RS, Al-Mouqdad MM. SMOFlipid Impact on Growth and Neonatal Morbidities in Very Preterm Infants. Nutrients 2022; 14:nu14193952. [PMID: 36235604 PMCID: PMC9573282 DOI: 10.3390/nu14193952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022] Open
Abstract
The soybean oil, medium-chain triglycerides, olive oil, and fish oil lipid (SMOFlipid) is increasingly being used worldwide without definite evidence of its benefits. We examined the effect of SMOFlipid on growth velocity and neonatal morbidities in very preterm infants. Very preterm infants who received soybean-based lipid emulsion between January 2015 and 2018 were compared with those who received SMOFlipids between 2019 and January 2022 in our neonatal tertiary center. Linear regression analysis was conducted to analyze the association between type of lipid emulsion and growth velocity. Modified log-Poisson regression with generalized linear models and a robust variance estimator (Huber−White) were applied to adjust for potential confounding factors. A total of 858 infants met our inclusion criteria. Of them, 238 (27.7%) received SMOFlipid. SMOFlipid was associated with lower growth velocity between birth and 36-week corrected gestational age compared with intralipid Δ weight z-score (adjusted mean difference (aMD) −0.67; 95% CI −0.69, −0.39). Subgroup analysis indicated that mainly male infants in the SMOFlipid−LE group had a lower Δ weight z-score compared to those in the intralipid group (p < 0.001), with no difference observed in females (p = 0.82). SMOFlipid was associated with a lower rate of bronchopulmonary dysplasia (BPD) (aRR 0.61; 95% CI 0.46, 0.8) and higher rate of late-onset sepsis compared with intralipid (aRR 1.44; 95% CI 1.22−1.69). SMOFlipid was associated with lower growth velocity and BPD but higher rate of late-onset sepsis—it is a double-edged sword.
Collapse
Affiliation(s)
- Suzan S. Asfour
- Clinical Pharmacy Department, Pharmaceutical Care Services, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Belal Alshaikh
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Latifah AlMahmoud
- Neonatal Intensive Care, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Haider H. Sumaily
- Neonatal Intensive Care, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Nabeel A. Alodhaidan
- Neonatal Intensive Care, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Mousa Alkhourmi
- Pediatric Gastroenterology Department, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Hissah A. Abahussain
- General Pediatrics Department, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Thanaa M. Khalil
- Obstetric and Gynecology Department, Maternity Hospital, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Bushra A. Albeshri
- Pharmacy Department, Pharmaceutical Care Services, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Aroub A. Alhamidi
- Pharmacy Department, Pharmaceutical Care Services, Ministry of Health, Riyadh 12613, Saudi Arabia
| | - Maha R. Al-Anazi
- Pharmacy Department, Pharmaceutical Care Services, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Raneem S. Asfour
- Pharmacy College, Jordan University of Science and Technology, Irbid P.O. Box 3030, Jordan
| | - Mountasser M. Al-Mouqdad
- Neonatal Intensive Care, Hospital of Pediatrics, King Saud Medical City, Riyadh 12746, Saudi Arabia
- Correspondence: ; Tel.: +966-114355555 (ext. 506); Fax: +966-114354406
| |
Collapse
|
39
|
Ryu J. New Aspects on the Treatment of Retinopathy of Prematurity: Currently Available Therapies and Emerging Novel Therapeutics. Int J Mol Sci 2022; 23:8529. [PMID: 35955664 PMCID: PMC9369302 DOI: 10.3390/ijms23158529] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/05/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a rare proliferative ocular disorder in preterm infants. Because of the advancements in neonatal care, the incidence of ROP has increased gradually. Now, ROP is one of the leading causes of blindness in children. Preterm infants with immature retinal development are exposed to supplemental oxygen inside an incubator until their cardiopulmonary system is adequately developed. Once they are returned to room air, the relatively low oxygen level stimulates various angiogenesis factors initiating retinal neovascularization. If patients with ROP are not offered adequate and timely treatment, they can experience vision loss that may ultimately lead to permanent blindness. Although laser therapy and anti-vascular endothelial growth factor agents are widely used to treat ROP, they have limitations. Thus, it is important to identify novel therapeutics with minimal adverse effects for the treatment of ROP. To date, various pharmacologic and non-pharmacologic therapies have been assessed as treatments for ROP. In this review, the major molecular factors involved in the pathogenesis of ROP, currently offered therapies, therapies under investigation, and emerging novel therapeutics of ROP are discussed.
Collapse
Affiliation(s)
- Juhee Ryu
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; ; Tel.: +82-539508583
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
40
|
Ferroptosis: A Promising Therapeutic Target for Neonatal Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2022; 23:ijms23137420. [PMID: 35806425 PMCID: PMC9267109 DOI: 10.3390/ijms23137420] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a type of programmed cell death caused by phospholipid peroxidation that has been implicated as a mechanism in several diseases resulting from ischemic-reperfusion injury. Most recently, ferroptosis has been identified as a possible key injury mechanism in neonatal hypoxic-ischemic brain injury (HIBI). This review summarizes the current literature regarding the different ferroptotic pathways, how they may be activated after neonatal HIBI, and which current or investigative interventions may attenuate ferroptotic cell death associated with neonatal HIBI.
Collapse
|
41
|
Mulder KA, Dyer RA, Elango R, Innis SM. Complexity of understanding the role of dietary and erythrocyte docosahexaenoic acid (DHA) on the cognitive performance of school-age children. Curr Dev Nutr 2022; 6:nzac099. [PMID: 35854937 PMCID: PMC9283108 DOI: 10.1093/cdn/nzac099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Early childhood is a period of rapid brain development, with increases in synapses rich in the omega-3 (ω-3) fatty acid, DHA (22:6ω-3) continuing well beyond infancy. Despite the importance of DHA to neural phospholipids, the requirement of dietary DHA for neurodevelopment remains unclear. Objectives The aim was to assess the dietary DHA and DHA status of young children, and determine the association with cognitive performance. Methods This was a cross-sectional study of healthy children (5-6 y), some of whom were enrolled in a follow-up of a clinical trial (NCT00620672). Dietary intake data (n = 285) were assessed with a food-frequency questionnaire (FFQ) and three 24-h recalls. Family characteristics were collected by questionnaire, and anthropometric data measured. Venous blood was collected, cognitive performance assessed using several age-appropriate tools including the Kaufman Assessment Battery for Children. The relation between dietary DHA, RBC DHA, and child neurodevelopment test scores was determined using Pearson's correlation or Spearman's rho, and quintiles of test scores compared by Mann-Whitney U test. Results Child DHA intakes were highly variable, with a stronger association between RBC DHA and DHA intake assessed by FFQ (rho = 0.383, P < 0.001) compared with one or three 24-h recalls. Observed ethnic differences in DHA intake status as well as neurodevelopmental test scores led to analysis of the association between DHA intake and status with neurodevelopment test scores for White children only (n = 190). Child RBC DHA status was associated with neurodevelopment test scores, including language (rho = 0.211, P = 0.009) and short-term memory (rho = 0.187, P = 0.019), but only short-term memory was associated with dietary DHA (rho = 0.221, P = 0.003). Conclusions Child RBC DHA but not dietary DHA was associated with multiple tests of cognitive performance. In addition, DHA intake was only moderately associated with RBC DHA, raising complex questions on the relation between diet, DHA transfer to membrane lipids, and neural function.
Collapse
Affiliation(s)
- Kelly A Mulder
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Roger A Dyer
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Analytical Core for Metabolics and Nutrition (ACMaN), BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Rajavel Elango
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Sheila M Innis
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
42
|
Henriksen NL, Hansen SH, Lycas MD, Pan X, Eriksen T, Johansen LS, Sprenger RR, Ejsing CS, Burrin DG, Skovgaard K, Christensen VB, Thymann T, Pankratova S. Cholestasis alters brain lipid and bile acid composition and compromises motor function in neonatal piglets. Physiol Rep 2022; 10:e15368. [PMID: 35822260 PMCID: PMC9277266 DOI: 10.14814/phy2.15368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/12/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022] Open
Abstract
Infants with neonatal cholestasis are prone to neurodevelopmental deficits, however, the underlying pathogenesis is unclear. Lipid malabsorption and accumulation of potentially neurotoxic molecules in the blood such as bile acids are important yet relatively unexplored pathways. Here, we developed a translational piglet model to understand how the molecular bile acid and lipid composition of the brain is affected by this disease and relates to motor function. Piglets (8-days old) had bile duct ligation or sham surgery and were fed a formula diet for 3 weeks. Alongside sensory-motor deficits observed in bile duct-ligated animals, we found a shift toward a more hydrophilic and conjugated bile acid profile in the brain. Additionally, comprehensive lipidomics of the cerebellum revealed a decrease in total lipids including phosphatidylinositols and phosphatidylserines and increases in lysophospholipid species. This was paralleled by elevated cerebellar expression of genes related to inflammation and tissue damage albeit without significant impact on the brain transcriptome. This study offers new insights into the developing brain's molecular response to neonatal cholestasis indicating that bile acids and lipids may contribute in mediating motor deficits.
Collapse
Affiliation(s)
- Nicole Lind Henriksen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Svend Høime Hansen
- Department of Clinical BiochemistryCopenhagen University Hospital, RigshospitaletCopenhagen ØDenmark
| | | | - Xiaoyu Pan
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Thomas Eriksen
- Department of Veterinary Clinical SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | | | - Richard R. Sprenger
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical SciencesUniversity of Southern DenmarkOdense MDenmark
| | - Christer Stenby Ejsing
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical SciencesUniversity of Southern DenmarkOdense MDenmark
| | - Douglas G. Burrin
- Department of Pediatrics, United States Department of Agriculture, Agricultural Research ServiceChildren's Nutrition Research Center, Baylor College of MedicineHoustonTexasUSA
| | - Kerstin Skovgaard
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Vibeke Brix Christensen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksberg CDenmark
- Department of Pediatrics and Adolescent MedicineCopenhagen University Hospital, RigshospitaletCopenhagen ØDenmark
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Stanislava Pankratova
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal SciencesUniversity of CopenhagenFrederiksberg CDenmark
| |
Collapse
|
43
|
Niwa S, Kawabata T, Shoji K, Ogata H, Kagawa Y, Nakayama K, Yanagisawa Y, Iwamoto S, Tatsuta N, Asato K, Arima T, Yaegashi N, Nakai K. Investigation of Maternal Diet and FADS1 Polymorphism Associated with Long-Chain Polyunsaturated Fatty Acid Compositions in Human Milk. Nutrients 2022; 14:nu14102160. [PMID: 35631303 PMCID: PMC9143760 DOI: 10.3390/nu14102160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
Increasing the amount of long-chain polyunsaturated fatty acids (LCPUFA) in human milk is an important strategy for infant growth and development. We investigated the associations of LCPUFA compositions in human milk with maternal diet (especially fish and shellfish intake), with fatty acid Δ5 desaturase gene (FADS1) polymorphisms, and with gene-diet interactions. The present study was performed as part of an adjunct study of the Japan Environment and Children’s Study. The participants were 304 lactating females, who provided human milk 6−7 months after delivery. Fatty acids in human milk were analyzed by gas chromatography, and dietary surveys were conducted using a brief self-administered diet history questionnaire. We also analyzed a single nucleotide polymorphism of FADS1 (rs174547, T/C). There was a significant difference in arachidonic acid (ARA) composition in human milk among the genotype groups, and the values were decreasing in the order of TT > TC > CC. The concentrations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were also different between TT and CC genotype, indicating a tendency for decreasing values in the same order. The composition of ARA showed significant gene−dietary interactions in multiple regression analysis, and the positive correlation between fish and shellfish intake and ARA composition in human milk was significant only in the CC genotype. Moreover, the factor most strongly associated with EPA and DHA composition in human milk was fish and shellfish intake. Therefore, it was suggested that increasing fish and shellfish intake in mothers may increase EPA and DHA composition in human milk, while increasing fish and shellfish intake in CC genotype mothers may lead to increased ARA composition in human milk.
Collapse
Affiliation(s)
- Sakurako Niwa
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
- Faculty of Home Economics, Gifu Women’s University, 80 Taromaru, Gifu City 501-2592, Gifu, Japan
- Correspondence: ; Tel.: +81-49-282-3705
| | - Terue Kawabata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Kumiko Shoji
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Hiromitsu Ogata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Yasuo Kagawa
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Kazuhiro Nakayama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8562, Chiba, Japan;
| | - Yoshiko Yanagisawa
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan; (Y.Y.); (S.I.)
| | - Sadahiko Iwamoto
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan; (Y.Y.); (S.I.)
| | - Nozomi Tatsuta
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
| | - Kaname Asato
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
| | - Takahiro Arima
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (T.A.); (N.Y.)
| | - Nobuo Yaegashi
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (T.A.); (N.Y.)
| | - Kunihiko Nakai
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
- School of Sport and Health Science, Tokai Gakuen University, Nishinohora 21-233, Miyoshi 470-0207, Aichi, Japan
| |
Collapse
|
44
|
Crawford MA, Wang Y, Marsh DE, Johnson MR, Ogundipe E, Ibrahim A, Rajkumar H, Kowsalya S, Kothapalli KSD, Brenna JT. Neurodevelopment, nutrition and genetics. A contemporary retrospective on neurocognitive health on the occasion of the 100th anniversary of the National Institute of Nutrition, Hyderabad, India. Prostaglandins Leukot Essent Fatty Acids 2022; 180:102427. [PMID: 35413515 PMCID: PMC9152880 DOI: 10.1016/j.plefa.2022.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
Abstract
In celebration of the centenary of the National Institute of Nutrition (NIN), Hyderabad, India (1918-2018), a symposium highlighted the progress in nutrition knowledge made over the century, as well as major gaps in implementation of that knowledge. Brain famine caused by a shortage of nutrients required for perinatal brain development has unfortunately become a global reality, even as protein-calorie famine was largely averted by the development of high yield crops. While malnutrition remains widespread, the neglect of global food policies that support brain development and maintenance are most alarming. Brain disorders now top the list of the global burden of disease, even with obesity rising throughout the world. Neurocognitive health, remarkably, is seldom listed among the non-communicable diseases (NCDs) and is therefore seldom considered as a component of food policy. Most notably, the health of mothers before conception and through pregnancy as mediated by proper nutrition has been neglected by the current focus on early death in non-neurocognitive NCDs, thereby compromising intellectual development of the ensuing generations. Foods with balanced essential fatty acids and ample absorbable micronutrients are plentiful for populations with access to shore-based foods, but deficient only a few kilometres away from the sea. Sustained access to brain supportive foods is a priority for India and throughout the world to enable each child to develop to their intellectual potential, and support a prosperous, just, and peaceful world. Nutrition education and food policy should place the nutritional requirements for the brain on top of the list of priorities.
Collapse
Affiliation(s)
- Michael A Crawford
- Institute of Brain Chemistry and Human Nutrition, Chelsea and Westminster Hospital Campus of Imperial College, London, United Kingdom
| | - Yiqun Wang
- Institute of Brain Chemistry and Human Nutrition, Chelsea and Westminster Hospital Campus of Imperial College, London, United Kingdom
| | - David E Marsh
- Institute of Brain Chemistry and Human Nutrition, Chelsea and Westminster Hospital Campus of Imperial College, London, United Kingdom
| | - Mark R Johnson
- Institute of Brain Chemistry and Human Nutrition, Chelsea and Westminster Hospital Campus of Imperial College, London, United Kingdom
| | - Enitan Ogundipe
- Institute of Brain Chemistry and Human Nutrition, Chelsea and Westminster Hospital Campus of Imperial College, London, United Kingdom
| | - Ahamed Ibrahim
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Hemalatha Rajkumar
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - S Kowsalya
- Department of Food Science and Nutrition, Avinashilingam Institute for Home Science and Higher Education for Women (Deemed to be University), Coimbatore, India
| | - Kumar S D Kothapalli
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, United States.
| | - J T Brenna
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, United States.
| |
Collapse
|
45
|
The protective effect of L-carnitine supplementation on retinopathy of prematurity: A retrospective cohort study. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.954325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
46
|
Fu Z, Yan W, Chen CT, Nilsson AK, Bull E, Allen W, Yang J, Ko M, SanGiovanni JP, Akula JD, Talukdar S, Hellström A, Smith LEH. Omega-3/Omega-6 Long-Chain Fatty Acid Imbalance in Phase I Retinopathy of Prematurity. Nutrients 2022; 14:1333. [PMID: 35405946 PMCID: PMC9002570 DOI: 10.3390/nu14071333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
There is a gap in understanding the effect of the essential ω-3 and ω-6 long-chain polyunsaturated fatty acids (LCPUFA) on Phase I retinopathy of prematurity (ROP), which precipitates proliferative ROP. Postnatal hyperglycemia contributes to Phase I ROP by delaying retinal vascularization. In mouse neonates with hyperglycemia-associated Phase I retinopathy, dietary ω-3 (vs. ω-6 LCPUFA) supplementation promoted retinal vessel development. However, ω-6 (vs. ω-3 LCPUFA) was also developmentally essential, promoting neuronal growth and metabolism as suggested by a strong metabolic shift in almost all types of retinal neuronal and glial cells identified with single-cell transcriptomics. Loss of adiponectin (APN) in mice (mimicking the low APN levels in Phase I ROP) decreased LCPUFA levels (including ω-3 and ω-6) in retinas under normoglycemic and hyperglycemic conditions. ω-3 (vs. ω-6) LCPUFA activated the APN pathway by increasing the circulating APN levels and inducing expression of the retinal APN receptor. Our findings suggested that both ω-3 and ω-6 LCPUFA are crucial in protecting against retinal neurovascular dysfunction in a Phase I ROP model; adequate ω-6 LCPUFA levels must be maintained in addition to ω-3 supplementation to prevent retinopathy. Activation of the APN pathway may further enhance the ω-3 and ω-6 LCPUFA's protection against ROP.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Wenjun Yan
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA;
| | - Chuck T. Chen
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20814, USA;
| | - Anders K. Nilsson
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 412 96 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Edward Bull
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - William Allen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Jay Yang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - John Paul SanGiovanni
- BIO5 Institute, Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA;
| | - James D. Akula
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Saswata Talukdar
- Cardiometabolic Diseases, Merck Research Laboratories, Boston, MA 02115, USA;
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 412 96 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Lois E. H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| |
Collapse
|
47
|
Firouzabadi FD, Shab-Bidar S, Jayedi A. The effects of omega-3 polyunsaturated fatty acids supplementation in pregnancy, lactation, and infancy: An umbrella review of meta-analyses of randomized trials. Pharmacol Res 2022; 177:106100. [PMID: 35104631 DOI: 10.1016/j.phrs.2022.106100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/12/2023]
Abstract
We aimed to perform an umbrella review of systematic reviews and meta-analyses (SRMAs) of randomized clinical trials (RCT) of the effects of long-chain omega-3 fatty acid supplementation in pregnancy, lactation, and infancy. We searched PubMed, Scopus, and Web of Science to November 2020. Two independent investigators extracted the information, evaluated the methodological quality of SRMAs using A Measurement Tool to Assess Systematic Reviews-2 (AMSTAR2), and rated the certainty of evidence using the Grades of Recommendation, Assessment, Development and Evaluation (GRADE) approach. Either a fixed-effects or a random-effects model was used to recalculate the effect sizes and 95%CIs, depending on the number of trials. Overall, 28 SRMAs of RCTs, reporting 124 outcomes from 672 RCTs with 273,523 participants were considered eligible for the present umbrella review. Our results demonstrated evidence of moderate to high certainty that omega-3 supplementation reduced the risk of pre-eclampsia and low-birth weight and improved head circumference when used in pregnant women, and reduced severe retinopathy of prematurity and cholestasis when used in infancy. There were also favorable effects on preterm delivery, pre-natal and post-partum depression, glycemic control and inflammation markers in pregnant women, and sensitization to peanuts, positive skin prick tests, anthropometric measures, language development, visual acuity, and duration of ventilation in infants (GRADE = low). Our findings suggested that omega-3 supplementation during pregnancy can exert favorable effects against pre-eclampsia, low-birth weight, pre-term delivery, and post-partum depression, and can improve anthropometric measures, immune system, and visual activity in infants and cardiometabolic risk factors in pregnant mothers.
Collapse
Affiliation(s)
- Fatemeh Dehghani Firouzabadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ahmad Jayedi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
48
|
Signorini C, De Felice C, Durand T, Galano JM, Oger C, Leoncini S, Hayek J, Lee JCY, Lund TC, Orchard PJ. Isoprostanoid Plasma Levels Are Relevant to Cerebral Adrenoleukodystrophy Disease. Life (Basel) 2022; 12:146. [PMID: 35207434 PMCID: PMC8874514 DOI: 10.3390/life12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebral adrenoleukodystrophy (ALD) is a rare neuroinflammatory disorder characterized by progressive demyelination. Mutations within the ABCD1 gene result in very long-chain fatty acid (VLCFA) accumulation within the peroxisome, particularly in the brain. While this VLCFA accumulation is known to be the driving cause of the disease, oxidative stress can be a contributing factor. For patients with early cerebral disease, allogeneic hematopoietic stem cell transplantation (HSCT) is the standard of care, and this can be supported by antioxidants. To evaluate the involvement of fatty acid oxidation in the disease, F2-isoprostanes (F2-IsoPs), F2-dihomo-isoprostanes (F2-dihomo-IsoPs) and F4-neuroprostanes (F4-NeuroPs)-which are oxygenated metabolites of arachidonic (ARA), adrenic (AdA) and docosahexaenoic (DHA) acids, respectively-in plasma samples from ALD subjects (n = 20)-with various phenotypes of the disease-were measured. Three ALD groups were classified according to patients with: (1) confirmed diagnosis of ALD but without cerebral disease; (2) cerebral disease in early period post-HSCT (<100 days post-HSCT) and on intravenous N-acetyl-L-cysteine (NAC) treatment; (3) cerebral disease in late period post-HSCT (beyond 100 days post-HSCT) and off NAC therapy. In our observation, when compared to healthy subjects (n = 29), in ALD (i), F2-IsoPs levels were significantly (p < 0.01) increased in all patients, with the single exception of the early ALD and on NAC subjects; (ii) significant elevated (p < 0.0001) amounts of F2-dihomo-IsoPs were detected, with the exception of patients with a lack of cerebral disease; (iii), a significant increase (p < 0.003) in F4-NeuroP plasma levels was detected in all ALD patients. Moreover, F2-IsoPs plasma levels were significantly higher (p = 0.038) in early ALD in comparison to late ALD stage, and F4-NeuroPs were significantly lower (p = 0.012) in ALD subjects with a lack of cerebral disease in comparison to the late disease stage. Remarkably, plasma amounts of all investigated isoprostanoids were shown to discriminate ALD patients vs. healthy subjects. Altogether, isoprostanoids are relevant to the phenotype of X-ALD and may be helpful in predicting the presence of cerebral disease and establishing the risk of progression.
Collapse
Affiliation(s)
- Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Claudio De Felice
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, CEDEX 5, 34093 Montpellier, France; (T.D.); (J.-M.G.); (C.O.)
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, CEDEX 5, 34093 Montpellier, France; (T.D.); (J.-M.G.); (C.O.)
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, CEDEX 5, 34093 Montpellier, France; (T.D.); (J.-M.G.); (C.O.)
| | - Silvia Leoncini
- Child Neuropsychiatry Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (S.L.); (J.H.)
| | - Joussef Hayek
- Child Neuropsychiatry Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (S.L.); (J.H.)
- Pediatric Speciality Center “L’Isola di Bau”, Certaldo, 50052 Florence, Italy
| | | | - Troy C. Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (T.C.L.); (P.J.O.)
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (T.C.L.); (P.J.O.)
| |
Collapse
|
49
|
Tardiff DF, Lucas M, Wrona I, Chang B, Chung CY, Le Bourdonnec B, Rhodes KJ, Scannevin RH. Non-clinical Pharmacology of YTX-7739: a Clinical Stage Stearoyl-CoA Desaturase Inhibitor Being Developed for Parkinson's Disease. Mol Neurobiol 2022; 59:2171-2189. [PMID: 35060064 PMCID: PMC9015998 DOI: 10.1007/s12035-021-02695-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/09/2021] [Indexed: 11/30/2022]
Abstract
Stearoyl-CoA desaturase (SCD) is a potential therapeutic target for Parkinson’s and related neurodegenerative diseases. SCD inhibition ameliorates neuronal toxicity caused by aberrant α-synuclein, a lipid-binding protein implicated in Parkinson’s disease. Its inhibition depletes monounsaturated fatty acids, which may modulate α-synuclein conformations and membrane interactions. Herein, we characterize the pharmacokinetic and pharmacodynamic properties of YTX-7739, a clinical-stage SCD inhibitor. Administration of YTX-7739 to rats and monkeys for 15 days caused a dose-dependent increase in YTX-7739 concentrations that were well-tolerated and associated with concentration-dependent reductions in the fatty acid desaturation index (FADI), the ratio of monounsaturated to saturated fatty acids. An approximate 50% maximal reduction in the carbon-16 desaturation index was observed in the brain, with comparable responses in the plasma and skin. A study with a diet supplemented in SCD products indicates that changes in brain C16 desaturation were due to local SCD inhibition, rather than to changes in systemic fatty acids that reach the brain. Assessment of pharmacodynamic response onset and reversibility kinetics indicated that approximately 7 days of dosing were required to achieve maximal responses, which persisted for at least 2 days after cessation of dosing. YTX-7739 thus achieved sufficient concentrations in the brain to inhibit SCD and produce pharmacodynamic responses that were well-tolerated in rats and monkeys. These results provide a framework for evaluating YTX-7739 pharmacology clinically as a disease-modifying therapy to treat synucleinopathies.
Collapse
Affiliation(s)
- Daniel F Tardiff
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.
| | - Matthew Lucas
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,Black Diamond Therapeutics, 1 Main Street, Cambridge, MA, 02142, USA
| | - Iwona Wrona
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,Black Diamond Therapeutics, 1 Main Street, Cambridge, MA, 02142, USA
| | - Belle Chang
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,iNeuro Therapeutics, 325 Vassar Street, Cambridge, MA, 02139, USA
| | - Chee Yeun Chung
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA
| | - Bertrand Le Bourdonnec
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,Deciphera Pharmaceuticals, 200 Smith St, Waltham, MA, 02451, USA
| | - Kenneth J Rhodes
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,Pfizer Rare Disease Research Unit, 1 Portland Street, Cambridge, MA, 02139, USA
| | - Robert H Scannevin
- Yumanity Therapeutics, 40 Guest Street, Suite 4410, Boston, MA, 02135, USA.,Verge Genomics, 2 Tower Pl, San Francisco, CA, 94080, USA
| |
Collapse
|
50
|
Heath RJ, Klevebro S, Wood TR. Maternal and Neonatal Polyunsaturated Fatty Acid Intake and Risk of Neurodevelopmental Impairment in Premature Infants. Int J Mol Sci 2022; 23:ijms23020700. [PMID: 35054885 PMCID: PMC8775484 DOI: 10.3390/ijms23020700] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
The N3 and N6 long chain polyunsaturated fatty acids (LCPUFA) docosahexaenoic acid (DHA) and arachidonic acid (AA) are essential for proper neurodevelopment in early life. These fatty acids are passed from mother to infant via the placenta, accreting into fetal tissues such as brain and adipose tissue. Placental transfer of LCPUFA is highest in the final trimester, but this transfer is abruptly severed with premature birth. As such, efforts have been made to supplement the post-natal feed of premature infants with LCPUFA to improve neurodevelopmental outcomes. This narrative review analyzes the current body of evidence pertinent to neurodevelopmental outcomes after LCPUFA supplementation in prematurely born infants, which was identified via the reference lists of systematic and narrative reviews and PubMed search engine results. This review finds that, while the evidence is weakened by heterogeneity, it may be seen that feed comprising 0.3% DHA and 0.6% AA is associated with more positive neurodevelopmental outcomes than LCPUFA-deplete feed. While no new RCTs have been performed since the most recent Cochrane meta-analysis in 2016, this narrative review provides a wider commentary; the wider effects of LCPUFA supplementation in prematurely born infants, the physiology of LCPUFA accretion into preterm tissues, and the physiological effects of LCPUFA that affect neurodevelopment. We also discuss the roles of maternal LCPUFA status as a modifiable factor affecting the risk of preterm birth and infant neurodevelopmental outcomes. To better understand the role of LCPUFAs in infant neurodevelopment, future study designs must consider absolute and relative availabilities of all LCPUFA species and incorporate the LCPUFA status of both mother and infant in pre- and postnatal periods.
Collapse
Affiliation(s)
- Rory J. Heath
- Emergency Medicine Department, Derriford Hospital, University Hospitals Plymouth NHS Foundation Trust, Plymouth PL68DH, UK;
| | - Susanna Klevebro
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, 11883 Stockholm, Sweden;
| | - Thomas R. Wood
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center on Human Development and Disability, University of Washington, Seattle, WA 98195, USA
- Institute for Human and Machine Cognition, Pensacola, FL 32502, USA
- Correspondence:
| |
Collapse
|