1
|
Farella I, D’Amato G, Orellana-Manzano A, Segura Y, Vitale R, Clodoveo ML, Corbo F, Faienza MF. "OMICS" in Human Milk: Focus on Biological Effects on Bone Homeostasis. Nutrients 2024; 16:3921. [PMID: 39599707 PMCID: PMC11597255 DOI: 10.3390/nu16223921] [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: 10/29/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Human milk (HM) is a complex biofluid rich in nutrients and bioactive compounds essential for infant health. Recent advances in omics technologies-such as proteomics, metabolomics, and transcriptomics-have shed light on the influence of HM on bone development and health. This review discusses the impact of various HM components, including proteins, lipids, carbohydrates, and hormones, on bone metabolism and skeletal growth. Proteins like casein and whey promote calcium absorption and osteoblast differentiation, supporting bone mineralization. Long-chain polyunsaturated fatty acids like docosahexaenoic acid (DHA) contribute to bone health by modulating inflammatory pathways and regulating osteoclast activity. Additionally, human milk oligosaccharides (HMOs) act as prebiotics, improving gut health and calcium bioavailability while influencing bone mineralization. Hormones present in HM, such as insulin-like growth factor 1 (IGF-1), leptin, and adiponectin, have been linked to infant growth, body composition, and bone density. Research has shown that higher IGF-1 levels in breast milk are associated with increased weight gain, while leptin and adiponectin influence fat mass and bone metabolism. Emerging studies have also highlighted the role of microRNAs (miRNAs) in regulating key processes like adipogenesis and bone homeostasis. Furthermore, microbiome-focused techniques reveal HM's role in establishing a balanced infant gut microbiota, indirectly influencing bone development by enhancing nutrient absorption. Although current findings are promising, comprehensive longitudinal studies integrating omics approaches are needed to fully understand the intricate relationships among maternal diet, HM composition, and infant bone health. Bridging these gaps could offer novel dietary strategies to optimize skeletal health during infancy, advancing early-life nutrition science.
Collapse
Affiliation(s)
- Ilaria Farella
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
| | - Gabriele D’Amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70012 Bari, Italy;
| | - Andrea Orellana-Manzano
- Escuela Superior Politécnica del Litoral, ESPOL, Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida (FCV), ESPOL Polytechnic University, Campus Gustavo Galindo Km 30.5 vía Perimetral, Guayaquil P.O. Box 09-01-5863, Ecuador; (A.O.-M.); (Y.S.)
| | - Yaritza Segura
- Escuela Superior Politécnica del Litoral, ESPOL, Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida (FCV), ESPOL Polytechnic University, Campus Gustavo Galindo Km 30.5 vía Perimetral, Guayaquil P.O. Box 09-01-5863, Ecuador; (A.O.-M.); (Y.S.)
| | - Rossella Vitale
- Giovanni XXIII Pediatric Hospital, University of Bari “A. Moro”, 70124 Bari, Italy;
| | - Maria Lisa Clodoveo
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari “A. Moro”, 70100 Bari, Italy;
| | - Filomena Corbo
- Department of Pharmacy-Drug Sciences, University of Bari “A. Moro”, 70125 Bari, Italy;
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A.Moro”, 70124 Bari, Italy
| |
Collapse
|
2
|
Kassaw AK, Bekele G, Kassaw AK, Yimer A. Prediction of acute respiratory infections using machine learning techniques in Amhara Region, Ethiopia. Sci Rep 2024; 14:27968. [PMID: 39543232 PMCID: PMC11564824 DOI: 10.1038/s41598-024-76847-3] [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: 12/19/2023] [Accepted: 10/17/2024] [Indexed: 11/17/2024] Open
Abstract
Many studies have shown that infectious diseases are responsible for the majority of deaths in children under five. Among these children, Acute Respiratory Infections is the most prevalent illness and cause of death worldwide. Acute respiratory infections continue to be the leading cause of death in developing countries, including Ethiopia. In order to predict the main factors contributing to acute respiratory infections in the Amhara regional state of Ethiopia, a machine learning technique was employed. This study utilized data from the 2016 Ethiopian Demographic and Health Survey. Seven machine learning models, including logistic regression, random forests, decision trees, Gradient Boosting, support vector machines, Naïve Bayes, and K-nearest neighbors, were employed to forecast the factors influencing acute respiratory infections. The accuracy of each model was assessed using receiver operating characteristic curves and various metrics. Among the seven models used, the Random Forest algorithm demonstrated the highest accuracy in predicting acute respiratory infections, with an accuracy rate of 90.35% and Area under the Curve of 94.80%. This was followed by the Decision Tree model with an accuracy rate of 88.69%, K-nearest neighbors with 86.35%, and Gradient Boosting with 82.69%. The Random Forest algorithm also exhibited positive and negative predictive values of 92.22% and 88.83%, respectively. Several factors were identified as significantly associated with ARI among children under five in the Amhara regional state, Ethiopia. These factors, included families with a poorer wealth status (log odds of 0.18) compared to their counterparts, families with four to six children (log odds of 0.1) compared to families with fewer than three living children, children without a history of diarrhea (log odds of -0.08), mothers who had occupation(log odds of 0.06) compared mothers who didn't have occupation, children under six months of age (log odds of -0.05) compared to children older than six months, mothers with no education (log odds of 0.04) compared to mothers with primary education or higher, rural residents (log odds of 0.03) compared to non-rural residents, families using wood as a cooking material (log odds of 0.03) compared to those using electricity. Through Shapley Additive exPlanations value analysis on the Random Forest algorithm, we have identified significant risk factors for acute respiratory infections among children in the Amhara regional state of Ethiopia. The study found that the family's wealth index, the number of children in the household, the mother's occupation, the mother's educational level, the type of residence, and the fuel type used for cooking were all associated with acute respiratory infections. Additionally, the research emphasized the importance of children being free from diarrhea and living in households with fewer children as essential factors for improving children's health outcomes in the Amhara regional state, Ethiopia.
Collapse
Affiliation(s)
- Abdulaziz Kebede Kassaw
- Department of Health Informatics, School of Public Health, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia.
| | - Gashaw Bekele
- Department of Software Engineering, College of Informatics, Kombolcha Institute of Technology, Wollo University, Kombolcha, Ethiopia
| | - Ahmed Kebede Kassaw
- Department of Information System, College of Informatics, Kombolcha Institute of Technology, Wollo University, Kombolcha, Ethiopia
| | - Ali Yimer
- Department of Public Health, College of Health Sciences, Woldia University, Woldia, Ethiopia
| |
Collapse
|
3
|
Yang Z, Li N, Cui H, Liu B, Wang X, Zhang L, Wang X, Zheng L, Yang X, Wu S, Hu J, Wen D. Influence of the Interaction between Genetic Factors and Breastfeeding on Children's Weight Status: A Systematic Review. Adv Nutr 2024; 15:100312. [PMID: 39389470 PMCID: PMC11566687 DOI: 10.1016/j.advnut.2024.100312] [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: 05/18/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
Breastfeeding may interact with other risk factors and have a combined influence on child growth. This systematic review aimed to examine the interaction between genetic factors and breastfeeding and how their combination is associated with children weight status. Four databases were searched until August 2024, and 8 eligible studies were identified. The fat mass and obesity associated (FTO) and peroxisome proliferator-activated receptor γ2 (PPARG2) genes were the most examined genes. Although the results of interactions between breastfeeding and genetics factors on children's weight status were inconsistent, some of studies reported that breastfeeding or exclusive breastfeeding attenuated the disadvantageous association between the risk alleles of the genes (higher obesity-specific genetic risk score for a multiple-gene study) and overdevelopment of children's body weight. These findings support the WHO recommendations for prolonged breastfeeding and further suggest breastfeeding interventions to prevent childhood obesity may be more effective in populations with a disadvantageous genetic predisposition. This review was registered in PROSPERO as CRD42023448365.
Collapse
Affiliation(s)
- Zhe Yang
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| | - Hong Cui
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| | - Borui Liu
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Xue Wang
- Medical Information Research Department/Library, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Xiaochuan Wang
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Lu Zheng
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Xinyue Yang
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Shuqi Wu
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Jiajin Hu
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China.
| | - Deliang Wen
- Health Sciences Institute, China Medical University, Shenyang, China; Research Center of China Medical University Birth Cohort, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Kilic A, Pehlivan S, Varkal MA, Tuncel FC, Kandemir I, Ozcetin M, Poyrazoglu S, Kardelen AD, Ozdemir I, Yildiz I. Investigating Leptin Gene Variants and Methylation Status in Relation to Breastfeeding and Preventing Obesity. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1293. [PMID: 39594868 PMCID: PMC11593199 DOI: 10.3390/children11111293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Objective: We investigated whether the results of leptin gene (LEP) 2548G/A (rs7799039) and leptin receptor gene (LEPR) 668 A/G (rs1137101) variants, as well as the methylation analysis of CpG regions at nucleotides -31 and -51 of the LEP gene, showed any differences between breastfed and non-breastfed children in this study. Materials and Methods: The cross-sectional study included 100 children aged 2-5 years who were attending nursery and kindergarten and had been accepted to the Department of General Paediatrics. Infants who were exclusively breastfed for the first six months after birth constituted the study group, and those who were not only breastfeed constituted the control group. Methylation percentages at CpG islands of the LEP gene were compared between exclusively breastfed and non-exclusively breastfed infants, and the statistical significance was analyzed by looking for changes in LEP -31 and -51 nt methylation and LEP 2548G/A ve LEPR 668 A/G variants. Results: Both groups were compared by feeding, and the association of LEPR and LEP gene polymorphisms and -51 nt and -31 nt methylations were analyzed. There were no significant differences between the groups regarding genotype and allele frequency for the LEPR 668 A/G, LEP 2548 G/A gene variant, -31 nt methylation, and -51 nt methylation status. Similarly, there was no significant difference in genotype and allele frequency for the LEPR 668 A/G gene variant in terms of duration of exclusive breastfeeding, total breastfeeding, body mass index, family obesity, and satiety status. However, maternal support from family elders and physical activity increased the 51 nt methylation, but this methylation was not significantly affected by BMI, age, or satiety status. Conclusions: Maternal support from family elders and physical activity were associated with increased 51 nt methylation, but this methylation was not significantly affected by BMI, age, or satiety status. However, there are not enough studies in this area to reach a definitive conclusion, and further research is needed.
Collapse
Affiliation(s)
- Ayse Kilic
- Division of General Pediatrics, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (A.K.); (M.A.V.); (I.Y.)
| | - Sacide Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (S.P.); (F.C.T.)
| | - Muhammet Ali Varkal
- Division of General Pediatrics, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (A.K.); (M.A.V.); (I.Y.)
| | - Fatima Ceren Tuncel
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (S.P.); (F.C.T.)
| | - Ibrahim Kandemir
- Department of Pediatrics, Faculty of Medicine, Istanbul Health and Technology University, 34445 Istanbul, Türkiye
| | - Mustafa Ozcetin
- Institute of Child Health, Istanbul University, 34093 Istanbul, Türkiye;
| | - Sükran Poyrazoglu
- Division of Pediatric Endocrinology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (S.P.); (A.D.K.)
| | - Asli Derya Kardelen
- Division of Pediatric Endocrinology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (S.P.); (A.D.K.)
| | - Irem Ozdemir
- Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye;
| | - Ismail Yildiz
- Division of General Pediatrics, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Türkiye; (A.K.); (M.A.V.); (I.Y.)
| |
Collapse
|
5
|
Melnik BC, Weiskirchen R, Stremmel W, John SM, Schmitz G. Risk of Fat Mass- and Obesity-Associated Gene-Dependent Obesogenic Programming by Formula Feeding Compared to Breastfeeding. Nutrients 2024; 16:2451. [PMID: 39125332 PMCID: PMC11314333 DOI: 10.3390/nu16152451] [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: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
It is the purpose of this review to compare differences in postnatal epigenetic programming at the level of DNA and RNA methylation and later obesity risk between infants receiving artificial formula feeding (FF) in contrast to natural breastfeeding (BF). FF bears the risk of aberrant epigenetic programming at the level of DNA methylation and enhances the expression of the RNA demethylase fat mass- and obesity-associated gene (FTO), pointing to further deviations in the RNA methylome. Based on a literature search through Web of Science, Google Scholar, and PubMed databases concerning the dietary and epigenetic factors influencing FTO gene and FTO protein expression and FTO activity, FTO's impact on postnatal adipogenic programming was investigated. Accumulated translational evidence underscores that total protein intake as well as tryptophan, kynurenine, branched-chain amino acids, milk exosomal miRNAs, NADP, and NADPH are crucial regulators modifying FTO gene expression and FTO activity. Increased FTO-mTORC1-S6K1 signaling may epigenetically suppress the WNT/β-catenin pathway, enhancing adipocyte precursor cell proliferation and adipogenesis. Formula-induced FTO-dependent alterations of the N6-methyladenosine (m6A) RNA methylome may represent novel unfavorable molecular events in the postnatal development of adipogenesis and obesity, necessitating further investigations. BF provides physiological epigenetic DNA and RNA regulation, a compelling reason to rely on BF.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Wolfgang Stremmel
- Praxis for Internal Medicine, Beethovenstrasse 2, D-76530 Baden-Baden, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
6
|
Jirillo E, Topi S, Charitos IA, Santacroce L, Gaxhja E, Colella M. Gut Microbiota and Immune System in Necrotizing Enterocolitis and Related Sepsis. GASTROINTESTINAL DISORDERS 2024; 6:431-445. [DOI: 10.3390/gidisord6020029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
A severe condition of sepsis can be a complication of necrotizing enterocolitis (NEC), which can occur in premature infants and becomes a medical challenge in the neonatal intensive care unit (NICU). It is a multifactorial intestinal disease (can affect both the small and large intestine) that can lead to ischemia of the intestinal tissues that evolves into acute organ necrosis. One of these factors is that different types of nutrition can influence the onset or the progression of the disease. Cow-milk-based infant formulas have been shown to cause it in premature infants more frequently than human milk. Recently, nutrition has been shown to be beneficial after surgery. Several issues still under study, such as the pathogenesis and the insufficient and often difficult therapeutic approach, as well as the lack of a common and effective prevention strategy, make this disease an enigma in daily clinical practice. Recent studies outlined the emerging role of the host immune system and resident gut microbiota, showing their close connection in NEC pathophysiology. In its initial stages, broad-spectrum antibiotics, bowel rest, and breastfeeding are currently used, as well as probiotics to help the development of the intestinal microbiota and its eubiosis. This paper aims to present the current knowledge and potential fields of research in NEC pathophysiology and therapeutic assessment.
Collapse
Affiliation(s)
- Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Elona Gaxhja
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
- Doctoral School, eCampus University, 22060 Novedrate, Italy
| |
Collapse
|
7
|
Qi Z, Zhang Z, Jin R, Zhang L, Zheng M, Li J, Wu Y, Li C, Lin B, Liu Y, Liu G. Target Analysis of Polychlorinated Naphthalenes and Nontarget Screening of Organic Chemicals in Bovine Milk, Infant Formula, and Adult Milk Powder by High-Resolution Mass Spectrometry. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:773-782. [PMID: 38109498 DOI: 10.1021/acs.jafc.3c07579] [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: 12/20/2023]
Abstract
Infant formula is intended as an effective substitute for breast milk but is the main source of polychlorinated naphthalenes (PCNs) to nonbreastfed infants. We performed target and nontarget analyses to determine PCNs and identify other organic contaminants in infant formula. The mean PCN concentrations in infant formula, milk powder, and bovine milk were 106.1, 88.8, and 78.2 μg kg-1 of dry weight, respectively. The PCN congener profiles indicated that thermal processes and raw materials were probably the main sources of PCNs in infant formula. A health risk assessment indicated that PCNs in infant formula do not pose health risks to infants. Using gas chromatography-Orbitrap mass spectrometry, 352, 372, and 161 organic chemicals were identified in the infant formula, milk powder, and bovine milk samples, respectively. Phthalate esters were detected in all four plastic-packed milk powder samples. The results indicated milk becomes more contaminated with organic chemicals during manufacturing, processing, and packaging.
Collapse
Affiliation(s)
- Ziyuan Qi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zherui Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rong Jin
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Lei Zhang
- China National Center for Food Safety Risk Assessment, Beijing 100021, P. R. China
| | - Minghui Zheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingguang Li
- China National Center for Food Safety Risk Assessment, Beijing 100021, P. R. China
| | - Yongning Wu
- China National Center for Food Safety Risk Assessment, Beijing 100021, P. R. China
| | - Cheng Li
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, P. R. China
| | - Bingcheng Lin
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Yahui Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Guorui Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Zurutuza JI, Gonzalez S, Calderón AL, Caba M, Ramos FR. Changes in the Immunology of Breast Milk From Obese or Overweight Women: a Brief Review. Cureus 2024; 16:e52207. [PMID: 38327967 PMCID: PMC10847007 DOI: 10.7759/cureus.52207] [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] [Accepted: 01/13/2024] [Indexed: 02/09/2024] Open
Abstract
A systematic search was carried out through search platforms and specialized databases, such as Academic Google, PubMed, and Scopus, using thesauri: breast feeding, obesity, immunology, and human milk in English and Spanish, and those articles published from January 2000 to December 2021, in both languages. Only those reports that included quantitative data on immunological components in the milk of normal-weight and overweight women were considered. The PRISMA 2020 guides were used, and a total of 306 articles were reviewed, of which a total of 33 were included, according to the basic inclusion criteria. It was observed that in obese mothers, there is an increase in certain immune cells, such as B lymphocytes, cytotoxic T lymphocytes, and NK cells, and cytokines, such as IL-6 and IFN-γ; other alterations included the bacterial population and proteins with antibacterial action. Also, a decrease in growth factors such as TGF-β and IFG-1 was documented in overweight women. Immunoglobulin concentrations did not show substantial changes. This brief review shows that maternal overweight is associated with changes in the biochemical and immunological parameters of milk.
Collapse
Affiliation(s)
- Jorge I Zurutuza
- Epidemiology and Biostatistics, Centro de Investigaciones Biomedicas, Universidad Veracruzana, Xalapa, MEX
| | - Santiago Gonzalez
- Chemistry, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, MEX
| | - Ana L Calderón
- Neuropathology, Instituto Nacional de Neurología y Neurocirugia, Ciudad de Mexico, MEX
| | - Mario Caba
- Neuroscience, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, MEX
| | - Fernando R Ramos
- Chemistry, Instituto de Quimica Aplicada, Universidad Veracruzana, Xalapa, MEX
| |
Collapse
|
9
|
Notarbartolo V, Carta M, Accomando S, Giuffrè M. The First 1000 Days of Life: How Changes in the Microbiota Can Influence Food Allergy Onset in Children. Nutrients 2023; 15:4014. [PMID: 37764797 PMCID: PMC10534753 DOI: 10.3390/nu15184014] [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: 08/04/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Allergic disease, including food allergies (FA)s, has been identified as a major global disease. The first 1000 days of life can be a "window of opportunity" or a "window of susceptibility", during which several factors can predispose children to FA development. Changes in the composition of the gut microbiota from pregnancy to infancy may play a pivotal role in this regard: some bacterial genera, such as Lactobacillus and Bifidobacterium, seem to be protective against FA development. On the contrary, Clostridium and Staphylococcus appear to be unprotective. METHODS We conducted research on the most recent literature (2013-2023) using the PubMed and Scopus databases. We included original papers, clinical trials, meta-analyses, and reviews in English. Case reports, series, and letters were excluded. RESULTS During pregnancy, the maternal diet can play a fundamental role in influencing the gut microbiota composition of newborns. After birth, human milk can promote the development of protective microbial species via human milk oligosaccharides (HMOs), which play a prebiotic role. Moreover, complementary feeding can modify the gut microbiota's composition. CONCLUSIONS The first two years of life are a critical period, during which several factors can increase the risk of FA development in genetically predisposed children.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Neonatal Intensive Care Unit with Neonatology, “G.F. Ingrassia” Hospital Unit, ASP 6, 90131 Palermo, Italy;
| | - Maurizio Carta
- Neonatology and Neonatal Intensive Care Unit, University Hospital Policlinic “Paolo Giaccone”, 90127 Palermo, Italy;
| | - Salvatore Accomando
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
| | - Mario Giuffrè
- Neonatology and Neonatal Intensive Care Unit, University Hospital Policlinic “Paolo Giaccone”, 90127 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
10
|
Abstract
Sialic acids (Sias), a group of over 50 structurally distinct acidic saccharides on the surface of all vertebrate cells, are neuraminic acid derivatives. They serve as glycan chain terminators in extracellular glycolipids and glycoproteins. In particular, Sias have significant implications in cell-to-cell as well as host-to-pathogen interactions and participate in various biological processes, including neurodevelopment, neurodegeneration, fertilization, and tumor migration. However, Sia is also present in some of our daily diets, particularly in conjugated form (sialoglycans), such as those in edible bird's nest, red meats, breast milk, bovine milk, and eggs. Among them, breast milk, especially colostrum, contains a high concentration of sialylated oligosaccharides. Numerous reviews have concentrated on the physiological function of Sia as a cellular component of the body and its relationship with the occurrence of diseases. However, the consumption of Sias through dietary sources exerts significant influence on human health, possibly by modulating the gut microbiota's composition and metabolism. In this review, we summarize the distribution, structure, and biological function of particular Sia-rich diets, including human milk, bovine milk, red meat, and egg.
Collapse
Affiliation(s)
- Tiantian Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jianrong Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiaobei Zhan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Wang X, Yan M, Zhang Y, Wang W, Zhang W, Luo J, Gan D, Yang H, Zhu S, He W. Breastfeeding in infancy and mortality in middle and late adulthood: A prospective cohort study and meta-analysis. J Intern Med 2023; 293:624-635. [PMID: 36815686 DOI: 10.1111/joim.13619] [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] [Indexed: 02/24/2023]
Abstract
BACKGROUND Breastfeeding in infancy is associated with a lower risk of mortality among children, but the impact on mortality in middle and late adulthood remains unknown. OBJECTIVES To assess the association between breastfeeding in infancy and mortality in middle and late adulthood. METHODS We included 383,627 participants aged 40-73 from the UK Biobank (2006-2010) and followed up until 2021. Hazard ratios (HRs) and 95% confidence intervals (CIs) for all-cause and cause-specific mortality according to breastfeeding in infancy were estimated with Cox proportional hazards regression models. We further did a meta-analysis, including results from our present study and three other cohort studies (PROSPERO; number CRD42022348925). RESULTS During a total of 4732,751 person-years of follow-up, 25,581 deaths were identified. Breastfeeding in infancy was associated with lower risks of mortality in middle and late adulthood, with adjusted HRs (95% CIs) of 0.95 (0.93-0.98) for all-cause mortality; 0.91 (0.87-0.96) for cardiovascular mortality and 0.94 (0.874-0.999) for respiratory mortality. Specifically, the association with mortality seemed to attenuate with age - stronger in middle-aged adults than in older adults. A similar association between breastfeeding in infancy and all-cause mortality was found in the meta-analysis. CONCLUSION Breastfeeding in infancy is associated with a lower risk of mortality - even decades later - in middle and late adulthood.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengsha Yan
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Yanyu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Wenjie Wang
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiwei Zhang
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junkai Luo
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Da Gan
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haomin Yang
- Department of Epidemiology and Health Statistics, School of Public Health & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shankuan Zhu
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei He
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Vázquez-Frias R, Ladino L, Bagés-Mesa MC, Hernández-Rosiles V, Ochoa-Ortiz E, Alomía M, Bejarano R, Boggio-Marzet C, Bojórquez-Ramos MC, Colindres-Campos E, Fernández G, García-Bacallao E, González-Cerda I, Guisande A, Guzmán C, Moraga-Mardones F, Palacios-Rosales J, Ramírez-Rodríguez NE, Roda J, Sanabria MC, Sánchez-Valverde F, Santiago RJ, Sepúlveda-Valbuena N, Spolidoro J, Valdivieso-Falcón P, Villalobos-Palencia N, Koletzko B. Consensus on complementary feeding from the Latin American Society for Pediatric Gastroenterology, Hepatology and Nutrition: COCO 2023. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2023; 88:57-70. [PMID: 36737343 DOI: 10.1016/j.rgmxen.2023.01.005] [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: 08/31/2022] [Accepted: 11/17/2022] [Indexed: 02/04/2023]
Abstract
Complementary feeding (CF) is defined as the feeding of infants that complements breastfeeding, or alternatively, feeding with a breast milk substitute, and is a process that is more than simply a guide as to what and how to introduce foods. The information provided by healthcare professionals must be up-to-date and evidence-based. Most of the recommendations that appear in the different international guidelines and position papers are widely applicable, but some must be regionalized or adapted to fit the conditions and reality of each geographic zone. The Nutrition Working Group of the Latin American Society for Pediatric Gastroenterology, Hepatology and Nutrition (LASPGHAN) summoned a group of experts from each of the society's member countries, to develop a consensus on CF, incorporating, whenever possible, local information adapted to the reality of the region. The aim of the present document is to show the results of that endeavor. Utilizing the Delphi method, a total of 34 statements on relevant aspects of CF were evaluated, discussed, and voted upon.
Collapse
Affiliation(s)
- R Vázquez-Frias
- Departamento de Gastroenterología y Nutrición, Instituto Nacional de Salud Hospital Infantil de México Federico Gómez, Mexico City, Mexico; Grupo de Nutrición, Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica, SLAGHNP/LASPGHAN.
| | - L Ladino
- Grupo de Nutrición, Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica, SLAGHNP/LASPGHAN; Facultad de Medicina, Grupo de Nutrición, Genética y Metabolismo, Universidad El Bosque, Bogotá, Colombia
| | - M C Bagés-Mesa
- Grupo de Nutrición, Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica, SLAGHNP/LASPGHAN; Facultad de Medicina, Grupo de Nutrición, Genética y Metabolismo, Universidad El Bosque, Bogotá, Colombia
| | - V Hernández-Rosiles
- Departamento de Gastroenterología y Nutrición, Instituto Nacional de Salud Hospital Infantil de México Federico Gómez, Mexico City, Mexico; Grupo de Nutrición, Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica, SLAGHNP/LASPGHAN
| | - E Ochoa-Ortiz
- Grupo de Nutrición, Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica, SLAGHNP/LASPGHAN; Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Departamento de Nutrición, Mexico City, Mexico
| | - M Alomía
- Posgrado de Pediatría, Facultad de Medicina, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - R Bejarano
- Servicio de Gastroenterología y Nutrición, Hospital de Especialidades Pediátricas, Ciudad de Panamá, Panama
| | - C Boggio-Marzet
- Grupo de Trabajo en Gastroenterología y Nutrición Pediátrica, Hospital General de Agudos "Dr. I. Pirovano", Buenos Aires, Argentina
| | | | - E Colindres-Campos
- Departamento de Pediatría, Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - G Fernández
- Departamento de Gastroenterología, Hospital Infantil Dr. Robert Reid Cabral, Santo Domingo, Dominican Republic
| | - E García-Bacallao
- Sección de Pediatría, Instituto de Gastroenterología, Universidad de Ciencias Médicas, La Habana, Cuba
| | - I González-Cerda
- Servicio de Pediatría, Hospital Militar Escuela "Dr. Alejandro Dávila Bolaños", Managua, Nicaragua
| | - A Guisande
- Centro Hospitalario Pereira Rossell, Montevideo, Uruguay
| | - C Guzmán
- Hospital La Católica, San José, Costa Rica
| | - F Moraga-Mardones
- Unidad de Nutrición, Servicio de Pediatría, Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - J Palacios-Rosales
- Facultad de Medicina, Universidad de San Carlos de Guatemala, Guatemala, Guatemala
| | - N E Ramírez-Rodríguez
- Facultad de Medicina, Departamento de Medicina y Salud Mental, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - J Roda
- Gastroenterología e Nutrição Pediátrica, Hospital Pediátrico - Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - M C Sanabria
- Cátedra y Servicio de Pediatría, Hospital de Clínicas, Facultad de Ciencias Médicas, Universidad Nacional de Asunción, Asunción, Paraguay
| | - F Sánchez-Valverde
- Sección de Gastroenterología y Nutrición Pediátrica, Hospital Universitario de Navarra, NAVARRA BIOMED, Pamplona, Navarra, Spain
| | - R J Santiago
- Departamento de Pediatría y Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario de Valera "Dr. Pedro Emilio Carrillo", Universidad de Los Andes, Valera, Venezuela
| | - N Sepúlveda-Valbuena
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - J Spolidoro
- Escuela de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - P Valdivieso-Falcón
- Departamento de Pediatría, Servicio subespecialidades pediátricas, Hospital Nacional Docente Madre Niño San Bartolomé, Lima, Peru
| | | | - B Koletzko
- Departamento de Pediatría, Medical Centre of LMU Munich, Ludwig Maximilian University of Munich, Dr. von Hauner Children's Hospital, München, Germany
| |
Collapse
|
13
|
Vázquez-Frias R, Ladino L, Bagés-Mesa M, Hernández-Rosiles V, Ochoa-Ortiz E, Alomía M, Bejarano R, Boggio-Marzet C, Bojórquez-Ramos M, Colindres-Campos E, Fernández G, García-Bacallao E, González-Cerda I, Guisande A, Guzmán C, Moraga-Mardones F, Palacios-Rosales J, Ramírez-Rodríguez N, Roda J, Sanabria M, Sánchez-Valverde F, Santiago R, Sepúlveda-Valbuena N, Spolidoro J, Valdivieso-Falcón P, Villalobos-Palencia N, Koletzko B. Consenso de alimentación complementaria de la Sociedad Latinoamericana de Gastroenterología, Hepatología y Nutrición Pediátrica: COCO 2023. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2023. [DOI: 10.1016/j.rgmx.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
14
|
Xu J, Shin J, McGee M, Unger S, Bando N, Sato J, Vandewouw M, Patel Y, Branson HM, Paus T, Pausova Z, O'Connor DL. Intake of mother's milk by very-low-birth-weight infants and variation in DNA methylation of genes involved in neurodevelopment at 5.5 years of age. Am J Clin Nutr 2022; 116:1038-1048. [PMID: 35977396 PMCID: PMC9535521 DOI: 10.1093/ajcn/nqac221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 08/09/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Mechanisms responsible for associations between intake of mother's milk in very-low-birth-weight (VLBW, <1500 g) infants and later neurodevelopment are poorly understood. It is proposed that early nutrition may affect neurodevelopmental pathways by altering gene expression through epigenetic modification. Variation in DNA methylation (DNAm) at cytosine-guanine dinucleotides (CpGs) is a commonly studied epigenetic modification. OBJECTIVES We aimed to assess whether early mother's milk intake by VLBW infants is associated with variations in DNAm at 5.5 y, and whether these variations correlate with neurodevelopmental phenotypes. METHODS This cohort study was a 5.5-y follow-up (2016-2018) of VLBW infants born in Ontario, Canada who participated in the Donor Milk for Improved Neurodevelopmental Outcomes trial. We performed an epigenome-wide association study (EWAS) to test whether percentage mother's milk (not including supplemental donor milk) during hospitalization was associated with DNAm in buccal cells during early childhood (n = 143; mean ± SD age: 5.7 ± 0.2 y; birth weight: 1008 ± 517 g). DNAm was assessed with the Illumina Infinium MethylationEPIC array at 814,583 CpGs. In secondary analyses, we tested associations between top-ranked CpGs and measures of early childhood neurodevelopment, e.g., total surface area of the cerebral cortex (n = 41, MRI) and Full-Scale IQ (n = 133, Wechsler Preschool and Primary Scale of Intelligence-IV). RESULTS EWAS analysis demonstrated percentage mother's milk intake by VLBW infants during hospitalization was associated with DNAm at 2 CpGs, cg03744440 [myosin XVB (MYO15B)] and cg00851389 [metallothionein 1A (MT1A)], at 5.5 y (P < 9E-08). Gene set enrichment analysis indicated that top-ranked CpGs (P < 0.001) were annotated to genes enriched in neurodevelopmental biological processes. Corroborating these findings, DNAm at several top identified CpGs from the EWAS was associated with cortical surface area and IQ at 5.5 y (P < 0.05). CONCLUSIONS In-hospital percentage mother's milk intake by VLBW infants was associated with variations in DNAm of neurodevelopmental genes at 5.5 y; some of these DNAm variations are associated with brain structure and IQ.This trial was registered at isrctn.com as ISRCTN35317141 and at clinicaltrials.gov as NCT02759809.
Collapse
Affiliation(s)
- Jingxiong Xu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Jean Shin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Meghan McGee
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, Sinai Health, Toronto, Ontario, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Bando
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Julie Sato
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Neuroscience & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marlee Vandewouw
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neuroscience & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Yash Patel
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Helen M Branson
- Division of Neuroradiology, Department of Medical Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Tomas Paus
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, Faculty of Medicine and CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine and CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Zdenka Pausova
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Deborah L O'Connor
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, Sinai Health, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Bottin JH, Eussen SRBM, Igbinijesu AJ, Mank M, Koyembi JCJ, Nyasenu YT, Ngaya G, Mad-Bondo D, Kongoma JB, Stahl B, Sansonetti PJ, Bourdet-Sicard R, Moya-Alvarez V. Food Insecurity and Maternal Diet Influence Human Milk Composition between the Infant's Birth and 6 Months after Birth in Central-Africa. Nutrients 2022; 14:4015. [PMID: 36235668 PMCID: PMC9573613 DOI: 10.3390/nu14194015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Although the World Health Organization (WHO) and UNICEF recommend that infants should be exclusively breastfed for the first 6 months of life, evidence is scarce on how the mother’s undernourishment status at delivery and maternal dietary factors influence human milk (HM) composition during the first 6 months of life in regions with high food insecurity. The maternal undernourishment status at delivery, maternal diet, and HM nutrients were assessed among 46 women and their 48 vaginally born infants in Bangui at 1, 4, 11, 18, and 25 weeks after birth through 24-h recalls and food consumption questionnaires from December 2017 to June 2019 in the context of the "Mother-to-Infant TransmIssion of microbiota in Central-Africa" (MITICA) study. High food insecurity indexes during the follow-up were significantly associated with them having lower levels of many of the human milk oligosaccharides (HMOs) that were measured and with lower levels of retinol (aß-coef = −0.2, p value = 0.04), fatty acids (aß-coef = −7.2, p value = 0.03), and amino acids (aß-coef = −2121.0, p value < 0.001). On the contrary, women from food-insecure households displayed significantly higher levels of lactose in their HM (aß-coef = 3.3, p value = 0.02). In parallel, the consumption of meat, poultry, and fish was associated with higher HM levels of many of the HMOs that were measured, total amino acids (aß-coef = 5484.4, p value < 0.001), and with lower HM levels of lactose (aß-coef = −15.6, p value = 0.01). Food insecurity and maternal diet had a meaningful effect on HM composition with a possible impact being an infant undernourishment risk. Our results plead for consistent actions on food security as an effective manner to influence the nutritional content of HM and thereby, potentially improve infant survival and healthy growth.
Collapse
Affiliation(s)
| | | | | | - Marko Mank
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands or
| | | | - Yawo Tufa Nyasenu
- Laboratoire d’Analyses Médicales, Institut Pasteur de Bangui, Bangui BP923, Central African Republic
- Laboratoire de Biologie Moléculaire et d’Immunologie, Université de Lomé, Lomé P.O. Box 1396, Togo
| | - Gilles Ngaya
- Laboratoire d’Analyses Médicales, Institut Pasteur de Bangui, Bangui BP923, Central African Republic
| | - Daniel Mad-Bondo
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri IZAMO, Bangui BP790, Central African Republic
| | - Jean-Bertrand Kongoma
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri IZAMO, Bangui BP790, Central African Republic
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands or
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Philippe J. Sansonetti
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, 75005 Paris, France
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, 75015 Paris, France
| | | | - Violeta Moya-Alvarez
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, 75015 Paris, France
- Epidemiology of Emergent Diseases Unit, Global Health Department, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
16
|
Bays HE, Golden A, Tondt J. Thirty Obesity Myths, Misunderstandings, and/or Oversimplifications: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2022. OBESITY PILLARS 2022; 3:100034. [PMID: 37990730 PMCID: PMC10661978 DOI: 10.1016/j.obpill.2022.100034] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2023]
Abstract
Background This Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) is intended to provide clinicians an overview of 30 common obesity myths, misunderstandings, and/or oversimplifications. Methods The scientific support for this CPS is based upon published citations, clinical perspectives of OMA authors, and peer review by the Obesity Medicine Association leadership. Results This CPS discusses 30 common obesity myths, misunderstandings, and/or oversimplifications, utilizing referenced scientific publications such as the integrative use of other published OMA CPSs to help explain the applicable physiology/pathophysiology. Conclusions This Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) on 30 common obesity myths, misunderstandings, and/or oversimplifications is one of a series of OMA CPSs designed to assist clinicians in the care of patients with the disease of obesity. Knowledge of the underlying science may assist the obesity medicine clinician improve the care of patients with obesity.
Collapse
Affiliation(s)
- Harold Edward Bays
- Louisville Metabolic and Atherosclerosis Research Center, University of Louisville School of Medicine, 3288, Illinois Avenue, Louisville, KY, 40213, USA
| | - Angela Golden
- NP Obesity Treatment Clinic, Flagstaff, AZ, 86001, USA
| | - Justin Tondt
- Department of Family and Community Medicine, Penn State Health, Penn State College of Medicine, 700 HMC Crescent Rd Hershey, PA, 17033, USA
| |
Collapse
|
17
|
Koemel NA, Skilton MR. Epigenetic Aging in Early Life: Role of Maternal and Early Childhood Nutrition. Curr Nutr Rep 2022; 11:318-328. [PMID: 35192186 PMCID: PMC9174131 DOI: 10.1007/s13668-022-00402-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Early life presents a pivotal period during which nutritional exposures are more likely to cause epigenetic modifications, which may impact an individual's health during adulthood. This article reviews the current evidence regarding maternal and early childhood nutritional exposures and their role in epigenetic aging. RECENT FINDINGS Maternal and early life consumption of diets higher in fiber, antioxidants, polyphenols, B vitamins, vitamin D, and ω-3 fatty acids is associated with slower epigenetic aging. Conversely, diets higher in glycemic load, fat, saturated fat, and ω-6 fatty acids demonstrate a positive association with epigenetic aging. Maternal and early life nutrition directly and indirectly influences epigenetic aging via changes in one-carbon metabolism, cardiometabolic health, and the microbiome. Clinical trials are warranted to determine the specific foods, dietary patterns, and dietary supplements that will normalize or lower epigenetic aging across the life course.
Collapse
Affiliation(s)
- Nicholas A. Koemel
- The Boden Initiative, Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michael R. Skilton
- The Boden Initiative, Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Sydney Institute for Women, Children and Their Families, Sydney Local Health District, Sydney, Australia
| |
Collapse
|
18
|
Ozkaya M, Korukcu O. Breast milk expression as a challenge for mothers of premature infants. J OBSTET GYNAECOL 2022; 42:1962-1971. [PMID: 35616235 DOI: 10.1080/01443615.2022.2055454] [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: 10/18/2022]
Abstract
The aim of this study was to examine the psychometric properties of the 11-item Breast Milk Expression Experience Scale and determine the level of breast milk expression experience of mothers after preterm delivery in Turkey. 165 mothers who agreed to participate between February 2019 and March 2020 used the Breast Milk Expression Experience Scale (BMEE-S). The general Cronbach alpha coefficient was 0.82 for the BMEE-S. The fit indices calculated by confirmatory factor analysis were χ2 (41) = 87.95, p = .00003, χ2/sd = 2.15, GFI = 0.91, NNFI = 0.93, CFI = 0.95, RMSEA = 0.08 and SRMR = 0.078. The BMEE-S three-factor structure was verified by the confirmatory factor analysis. The BMEE-S was a valid and reliable instrument for mothers of preterm infants. Multiparity and high-risk pregnancy history negatively affected the milk expression experience, whereas spousal support positively affected it.Impact statementWhat is already known on this subject? The risk of infectious diseases, obesity, diabetes, and impaired intellectual development increases in children who do not breast milk (Verduci et al. 2014). International breastfeeding guidelines recommend supporting all mothers to start breastfeeding within one hour of birth, and counselling mothers who are separated from their babies about expressing milk.What the results of this study add? The breast milk expression experience scale is a valid and reliable tool for women who give preterm birth and express milk. The experience of expressing breast milk can affect the amount of milk that passes from mother to baby. Multiparity and high risk pregnancy history of mothers negatively affects the milk expression experience while husband support positively affects the milk expression experience.What the implications are of these findings for clinical practice and/or further research? Our study provides a quantitative examination of the experiences of mothers who are separated from their babies. The BMEE-S should be adapted to different cultures and the experiences of mothers in this process should be determined.
Collapse
Affiliation(s)
- Meltem Ozkaya
- Faculty of Nursing, Akdeniz University, Antalya, Turkey
| | - Oznur Korukcu
- Faculty of Nursing, Akdeniz University, Antalya, Turkey
| |
Collapse
|
19
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
20
|
Using complexity science to understand the role of co-sleeping (bedsharing) in mother-infant co-regulatory processes. Infant Behav Dev 2022; 67:101723. [PMID: 35594598 DOI: 10.1016/j.infbeh.2022.101723] [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: 05/02/2021] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022]
Abstract
Human infants spend most of their time sleeping, but over the first few years of life their sleep becomes regulated to coincide more closely with adult sleep (Galland et al., 2012; Paavonen et al., 2020). Evidence shows that co-sleeping played a role in the evolution of infant sleep regulation, as it is part of an ancient behavioral complex representing the biopsychosocial microenvironment in which human infants co-evolved with their mothers through millions of years of human history (Ball, 2003; McKenna 1986, 1990). This paper is a conceptual, interdisciplinary, integration of the literature on mother-infant co-sleeping and other mother-infant co-regulatory processes from an evolutionary (biological) perspective, using complexity science. Viewing the mother-infant dyad as a "complex adaptive system" (CAS) shows how the CAS fits assumptions of regulatory processes and reveals the role of the CAS in the ontogeny of mother-infant co-regulation of physiological (thermoregulation, breathing, circadian rhythm coordination, nighttime synchrony, and heart rate variability) and socioemotional (attachment and cortisol activity) development.
Collapse
|
21
|
Nutrition during Pregnancy and Lactation: Epigenetic Effects on Infants’ Immune System in Food Allergy. Nutrients 2022; 14:nu14091766. [PMID: 35565735 PMCID: PMC9103859 DOI: 10.3390/nu14091766] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
Food allergies are an increasing health problem worldwide. They are multifactorial diseases, in which the genome alone does not explain the development of the disease, but a genetic predisposition and various environmental factors contribute to their onset. Environmental factors, in particular nutritional factors, in the early stages of life are recognized as key elements in the etiology of food allergies. There is growing evidence advising that nutrition can affect the risk of developing food allergies through epigenetic mechanisms elicited by the nutritional factors themselves or by modulating the gut microbiota and its functional products. Gut microbiota and postbiotics can in turn influence the risk of food allergy development through epigenetic mechanisms. Epigenetic programming accounts not only for the short-term effects on the individual’s health status, but also for those observed in adulthood. The first thousand days of life represent an important window of susceptibility in which environmental factors, including nutritional ones, can influence the risk of developing allergies through epigenetic mechanisms. From this point of view, it represents an interesting window of opportunity and intervention. This review reports the main nutritional factors that in the early stages of life can influence immune oral tolerance through the modulation of epigenetic mechanisms.
Collapse
|
22
|
Nutritional Composition of Traditional Complementary Foods in Nigeria and Health / Developmental Outcomes: A Systematic Review. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Duale A, Singh P, Al Khodor S. Breast Milk: A Meal Worth Having. Front Nutr 2022; 8:800927. [PMID: 35155521 PMCID: PMC8826470 DOI: 10.3389/fnut.2021.800927] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
A mother is gifted with breast milk, the natural source of nutrition for her infant. In addition to the wealth of macro and micro-nutrients, human milk also contains many microorganisms, few of which originate from the mother, while others are acquired from the mouth of the infant and the surroundings. Among these microbes, the most commonly residing bacteria are Staphylococci, Streptococci, Lactobacilli and Bifidobacteria. These microorganisms initiate and help the development of the milk microbiota as well as the microbiota of the gastrointestinal tract in infants, and contribute to developing immune regulatory factors such as cytokines, growth factors, lactoferrin among others. These factors play an important role in reducing the risk of developing chronic diseases like type 2 diabetes, asthma and others later in life. In this review, we will summarize the known benefits of breastfeeding and highlight the role of the breast milk microbiota and its cross-talk with the immune system in breastfed babies during the early years of life.
Collapse
Affiliation(s)
- Anoud Duale
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Parul Singh
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Ar-Rayyan, Qatar
| | - Souhaila Al Khodor
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
- *Correspondence: Souhaila Al Khodor
| |
Collapse
|
24
|
Wallenborn JT, Gunier RB, Pappas DJ, Chevrier J, Eskenazi B. Breastmilk, Stool, and Meconium: Bacterial Communities in South Africa. MICROBIAL ECOLOGY 2022; 83:246-251. [PMID: 33885917 PMCID: PMC8531170 DOI: 10.1007/s00248-021-01758-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/18/2021] [Indexed: 06/12/2023]
Abstract
Human milk optimizes gut microbial richness and diversity, and is critical for proper immune development. Research has shown differing microbial composition based on geographic location, providing evidence that diverse biospecimen data is needed when studying human bacterial communities. Yet, limited research describes human milk and infant gut microbial communities in Africa. Our study uses breastmilk, stool, and meconium samples from a South African birth cohort to describe the microbial diversity, identify distinct taxonomic units, and determine correlations between bacterial abundance in breastmilk and stool samples. Mother-infant dyads (N = 20) were identified from a longitudinal birth cohort in the Vhembe district of Limpopo Province, South Africa. Breastmilk, meconium, and stool samples were analyzed using 16S ribosomal RNA sequencing of the V4-V5 gene region using the MiSeq platform for identification and relative quantification of bacterial taxa. A non-metric multidimensional scaling using Bray-Curtis distances of sample Z-scores showed that meconium, stool, and breastmilk microbial communities are distinct with varying genus. Breastmilk was mostly comprised of Streptococcus, Staphylococcus, Veillonella, and Corynebacterium. Stool samples showed the highest levels of Bifidobacterium, Faecalibacterium, Bacteroides, and Streptococcus. Alpha diversity measures found that stool samples have the highest Shannon index score compared to breastmilk and meconium. The abundance of Bifidobacterium (r = 0.57), Blautia (r = 0.59), and Haemophilus (r = 0.69) was correlated (p < 0.1) between breastmilk and stool samples. Despite the importance of breastmilk in seeding the infant gut microbiome, we found evidence of distinct bacterial communities between breastmilk and stool samples from South African mother-infant dyads.
Collapse
Affiliation(s)
- Jordyn T Wallenborn
- Maternal and Child Health Program, School of Public Health, University of California Berkeley, 1995 University Ave, Suite 265, Berkeley, CA, 94704, USA.
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland.
| | - Robert B Gunier
- Center for Environmental Research and Children's Health, University of California Berkeley, Berkeley, CA, USA
| | - Derek J Pappas
- California Institute for Quantitative Biosciences, Genomics Sequencing Laboratory, University of California Berkeley, Berkeley, CA, USA
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health, University of California Berkeley, Berkeley, CA, USA
| |
Collapse
|
25
|
Briollais L, Rustand D, Allard C, Wu Y, Xu J, Rajan SG, Hivert MF, Doyon M, Bouchard L, McGowan PO, Matthews S, Lye S. DNA methylation mediates the association between breastfeeding and early-life growth trajectories. Clin Epigenetics 2021; 13:231. [PMID: 34937578 PMCID: PMC8697471 DOI: 10.1186/s13148-021-01209-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/04/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The role of breastfeeding in modulating epigenetic factors has been suggested as a possible mechanism conferring its benefits on child development but it lacks evidence. Using extensive DNA methylation data from the ALSPAC child cohort, we characterized the genome-wide landscape of DNA methylation variations associated with the duration of exclusive breastfeeding and assessed whether these variations mediate the association between exclusive breastfeeding and BMI over different epochs of child growth. RESULTS Exclusive breastfeeding elicits more substantial DNA methylation variations during infancy than at other periods of child growth. At the genome-wide level, 13 CpG sites in girls (miR-21, SNAPC3, ATP6V0A1, DHX15/PPARGC1A, LINC00398/ALOX5AP, FAM238C, NATP/NAT2, CUX1, TRAPPC9, OSBPL1A, ZNF185, FAM84A, PDPK1) and 2 CpG sites in boys (IL16 and NREP), mediate the association between exclusive breastfeeding and longitudinal BMI. We found enrichment of CpG sites located within miRNAs and key pathways (AMPK signaling pathway, insulin signaling pathway, endocytosis). Overall DNA methylation variation corresponding to 3 to 5 months of exclusive breastfeeding was associated with slower BMI growth the first 6 years of life compared to no breastfeeding and in a dose-response manner with exclusive breastfeeding duration. CONCLUSIONS Our study confirmed the early postnatal period as a critical developmental period associated with substantial DNA methylation variations, which in turn could mitigate the development of overweight and obesity from infancy to early childhood. Since an accelerated growth during these developmental periods has been linked to the development of sustained obesity later in life, exclusive breastfeeding could have a major role in preventing the risks of overweight/obesity and children and adults through DNA methylation mechanisms occurring early in life.
Collapse
Affiliation(s)
- Laurent Briollais
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 60, Murray street - Room 5-237, Toronto, ON, M5T 3L9, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| | - Denis Rustand
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 60, Murray street - Room 5-237, Toronto, ON, M5T 3L9, Canada
- Biostatistics Team, Bordeaux Population Health Center, ISPED, Centre INSERM U1219, Bordeaux, France
| | - Catherine Allard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Yanyan Wu
- Department of Public Health Sciences, University of Hawai'i at Manoa, Honolulu, HI, USA
| | - Jingxiong Xu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 60, Murray street - Room 5-237, Toronto, ON, M5T 3L9, Canada
| | | | - Marie-France Hivert
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, 02215, USA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Myriam Doyon
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Luigi Bouchard
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean, Hôpital Universitaire de Chicoutimi, Saguenay, QC, G7H 5H6, Canada
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Patrick O McGowan
- Department of Biological Sciences, University of Toronto - Scarborough, Toronto, ON, Canada
| | - Steven Matthews
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 60, Murray street - Room 5-237, Toronto, ON, M5T 3L9, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Steven Lye
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 60, Murray street - Room 5-237, Toronto, ON, M5T 3L9, Canada
| |
Collapse
|
26
|
Polverino A, Sorrentino P, Pesoli M, Mandolesi L. Nutrition and cognition across the lifetime: an overview on epigenetic mechanisms. AIMS Neurosci 2021; 8:448-476. [PMID: 34877399 PMCID: PMC8611190 DOI: 10.3934/neuroscience.2021024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/12/2021] [Indexed: 12/28/2022] Open
Abstract
The functioning of our brain depends on both genes and their interactions with environmental factors. The close link between genetics and environmental factors produces structural and functional cerebral changes early on in life. Understanding the weight of environmental factors in modulating neuroplasticity phenomena and cognitive functioning is relevant for potential interventions. Among these, nutrition plays a key role. In fact, the link between gut and brain (the gut-brain axis) is very close and begins in utero, since the Central Nervous System (CNS) and the Enteric Nervous System (ENS) originate from the same germ layer during the embryogenesis. Here, we investigate the epigenetic mechanisms induced by some nutrients on the cognitive functioning, which affect the cellular and molecular processes governing our cognitive functions. Furthermore, epigenetic phenomena can be positively affected by specific healthy nutrients from diet, with the possibility of preventing or modulating cognitive impairments. Specifically, we described the effects of several nutrients on diet-dependent epigenetic processes, in particular DNA methylation and histones post-translational modifications, and their potential role as therapeutic target, to describe how some forms of cognitive decline could be prevented or modulated from the early stages of life.
Collapse
Affiliation(s)
- Arianna Polverino
- Institute of Diagnosis and Treatment Hermitage Capodimonte, Naples, Italy.,Department of Motor and Wellness Sciences, University of Naples "Parthenope", Naples, Italy
| | - Pierpaolo Sorrentino
- Institut de Neurosciences des Systèmes, Aix-Marseille University, Marseille, France.,Institute of Applied Sciences and Intelligent Systems, National Research Council, Pozzuoli, Italy
| | - Matteo Pesoli
- Department of Motor and Wellness Sciences, University of Naples "Parthenope", Naples, Italy
| | - Laura Mandolesi
- Department of Humanities Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
27
|
de Castilhos JK, Campagnolo PDB, de Almeida S, Vitolo MR, Mattevi VS. Impact of maternal dietary counseling in the first year of life on DNA methylation in a cohort of children. Genet Mol Biol 2021; 44:e20200330. [PMID: 34874049 PMCID: PMC8670011 DOI: 10.1590/1678-4685-gmb-2020-0330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/28/2021] [Indexed: 01/16/2023] Open
Abstract
Epigenetic modifications established during prenatal and early life, including DNA methylation, have been suggested as potential mediators of the interaction between environmental exposures during the perinatal period and adult metabolic health adverse outcomes, especially cardiometabolic complications and overweight. The effect of a dietary intervention in the first year of life on global methylation levels in leukocyte samples from a cohort of children born between 2001 and 2002 in southern Brazil was examined. Overall methylation measurements were performed using enzyme-linked immunosorbent assays on DNA samples from 237 children at 4 years old. Mean methylation values were higher in the intervention group (mean: 2.20 ± 1.31%) than in the control group (mean: 1.65 ± 1.11%; P = 0.001). It was observed that nutritional counseling in the first year increased breastfeeding duration and stimulated the development of healthier eating habits. Therefore, these factors might have contributed to increase global DNA methylation. The findings of the present study reinforce the notion that performing nutritional interventions in the early stages of life is important and provide further evidence of the interaction between the environment and epigenetic traits.
Collapse
Affiliation(s)
| | | | - Silvana de Almeida
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Márcia Regina Vitolo
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Vanessa Suñé Mattevi
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
28
|
Chaudron Y, Pifferi F, Aujard F. Overview of age-related changes in psychomotor and cognitive functions in a prosimian primate, the gray mouse lemur (Microcebus murinus): Recent advances in risk factors and antiaging interventions. Am J Primatol 2021; 83:e23337. [PMID: 34706117 DOI: 10.1002/ajp.23337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 01/13/2023]
Abstract
Aging is not homogeneous in humans and the determinants leading to differences between subjects are not fully understood. Impaired glucose homeostasis is a major risk factor for cognitive decline in middle-aged humans, pointing at the existence of early markers of unhealthy aging. The gray mouse lemur (Microcebus murinus), a small lemuriform Malagasy primate, shows relatively slow aging with decreased psychomotor capacities at middle-age (around 5-year old). In some cases (∼10%), it spontaneously leads to pathological aging. In this case, some age-related deficits, such as severe cognitive decline, brain atrophy, amyloidosis, and glucoregulatory imbalance are congruent with what is observed in humans. In the present review, we inventory the changes occurring in psychomotor and cognitive functions during healthy and pathological aging in mouse lemur. It includes a summary of the cerebral, metabolic, and cellular alterations that occur during aging and their relation to cognitive decline. As nutrition is one of the major nonpharmacological antiaging strategies with major potential effects on cognitive performances, we also discuss its role in brain functions and cognitive decline in this species. We show that the overall approach of aging studies in the gray mouse lemur offers promising ways of investigation for understanding, prevention, and treatments of pathological aging in humans.
Collapse
Affiliation(s)
- Yohann Chaudron
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Fabien Pifferi
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Fabienne Aujard
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| |
Collapse
|
29
|
Fiocchi A, Knol J, Koletzko S, O’Mahony L, Papadopoulos NG, Salminen S, Szajewska H, Nowak-Węgrzyn A. Early-Life Respiratory Infections in Infants with Cow's Milk Allergy: An Expert Opinion on the Available Evidence and Recommendations for Future Research. Nutrients 2021; 13:3795. [PMID: 34836050 PMCID: PMC8621023 DOI: 10.3390/nu13113795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Acute respiratory infections are a common cause of morbidity in infants and young children. This high rate of respiratory infections in early life has a major impact on healthcare resources and antibiotic use, with the associated risk of increasing antibiotic resistance, changes in intestinal microbiota composition and activity and, consequently, on the future health of children. An international group of clinicians and researchers working in infant nutrition and cow's milk allergy (CMA) met to review the available evidence on the prevalence of infections in healthy infants and in those with allergies, particularly CMA; the factors that influence susceptibility to infection in early life; links between infant feeding, CMA and infection risk; and potential strategies to modulate the gut microbiota and infection outcomes. The increased susceptibility of infants with CMA to infections, and the reported potential benefits with prebiotics, probiotics and synbiotics with regard to improving infection outcomes and reducing antibiotic usage in infants with CMA, makes this a clinically important issue that merits further research.
Collapse
Affiliation(s)
- Alessandro Fiocchi
- Translational Research in Pediatric Specialities Area, Division of Allergy, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
- The Laboratory of Microbiology, Wageningen University, 6700 HB Wageningen, The Netherlands
| | - Sibylle Koletzko
- Dr von Hauner Kinderspital, University Hospital, LMU Klinikum, 80337 Munich, Germany;
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, 10-719 Olsztyn, Poland
| | - Liam O’Mahony
- Department of Medicine, School of Microbiology, APC Microbiome Ireland National University of Ireland, T12 K8AF Cork, Ireland;
| | - Nikolaos G. Papadopoulos
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9WL, UK;
- Allergy Department, 2nd Pediatric Clinic, University of Athens, 11527 Athens, Greece
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20014 Turku, Finland;
| | - Hania Szajewska
- Department of Paediatrics, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Anna Nowak-Węgrzyn
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, 10-719 Olsztyn, Poland
- Department of Pediatrics, NYU Grossman School of Medicine, Hassenfeld Children’s Hospital, New York, NY 10016, USA
| |
Collapse
|
30
|
Windi R, Efendi F, Qona'ah A, Adnani QES, Ramadhan K, Almutairi WM. Determinants of Acute Respiratory Infection Among Children Under-Five Years in Indonesia. J Pediatr Nurs 2021; 60:e54-e59. [PMID: 33744057 DOI: 10.1016/j.pedn.2021.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute respiratory infection (ARI) among children under five years has been identified as a risk factor for child morbidity, leading to child mortality in Indonesia. Many factors may cause ARI; however, determinants associated with ARI remain unclear in Indonesia. OBJECTIVES This study sought to analyze the determinants of ARI among children aged under five years in Indonesia. METHODS This study was cross-sectional and utilized secondary data from the 2017 Indonesian Demographic and Health Survey (IDHS). A total of 15,993 children under five years old were selected as respondents. Chi-squared test and binary logistic regression were used to examine the determinants of ARI among children under five years in Indonesia. RESULTS Children aged 1 year [Odds Ratio (OR) = 1.43, 95% CI = 1.04-1.97], children aged 2 years [OR = 1.54, 95% CI = 1.12-2.11], mother's occupation [OR = 1.24, 95% CI = 1.01-2.154], poorest wealth index [OR = 1.91, 95% CI = 1.26-2.89], poor [OR = 1.50, 95% CI = 1.01-2.21], region of residence: Western Indonesia [OR = 1.96, 95% CI = 1.28-2.00], Middle of Indonesia [OR = 2.19, 95% CI = 1.44-3.33] were significantly associated with ARI among children under five years in Indonesia. CONCLUSIONS This study revealed that the determinants of ARI among children under five years in Indonesia remain related to the socio-demographic aspect. This research highlighted that the family's and the living area's wealth index remains essential in improving children's health outcomes. PRACTICE IMPLICATIONS Our findings support increasing awareness of the low-income family through adequate information and health promotion. Advancing the feasibility, accessibility, and affordability of health information and health services across all Indonesian regions should be strengthened.
Collapse
Affiliation(s)
- Restu Windi
- Faculty of Nursing, Universitas Airlangga, Indonesia
| | - Ferry Efendi
- Faculty of Nursing, Universitas Airlangga, Indonesia.
| | - Arina Qona'ah
- Faculty of Nursing, Universitas Airlangga, Indonesia.
| | | | - Kadar Ramadhan
- Department of Midwifery, Poltekkes Kemekes Palu, Indonesia
| | - Wedad M Almutairi
- Department of Maternity and Childhood, Faculty of Nursing, King Abdulaziz University, Saudi Arabia.
| |
Collapse
|
31
|
Molecular epigenetic dynamics in breast carcinogenesis. Arch Pharm Res 2021; 44:741-763. [PMID: 34392501 DOI: 10.1007/s12272-021-01348-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer has become one of the most common dreadful diseases that target women across the globe. The most obvious reasons we associate with it are either genetic mutations or dysregulation of pathways. However, there is yet another domain that has a significant role in influencing the genetic mutations and pathways. Epigenetic mechanisms influence these pathways either independently or in association with genetic mutations, thereby expediting the process of breast carcinogenesis. Breast cancer is governed by various transduction pathways such as PI3K/AKT/mTOR, NOTCH, β Catenin, NF-kB, Hedgehog, etc. There are many proteins as well that serve to be tumor suppressors but somehow lose their ability to function. This may be because of either genetic mutation or a process that represses their function. Apart from these, there are a lot of individual factors like puberty, breastfeeding, abortion, parity, circadian rhythm, alcohol consumption, pollutants, and obesity that drive these mutations and hence alter the pathways. Epigenetic mechanisms like DNA methylation, histone modifications, and lncRNAs directly or indirectly bring alterations in the proteins that are involved in the pathways. They do this by either promoting the transcription of genes or by repressing it at the ground genetic level that advances breast carcinogenesis. Epigenetics precedes genetic mutation in driving carcinogenesis and so, it needs to be explored further to diversify the possibilities of target specific treatments. In this review, the general role of DNA methylation, histone modification, and lncRNAs in breast cancer and their role in influencing the oncogenic signaling pathways along with the various factors governing them have been discussed for a better understanding of the role of epigenetics in breast carcinogenesis.
Collapse
|
32
|
Silva MDB, de Oliveira RDVC, Alves DDSB, Melo ECP. The effect of risk at birth on breastfeeding duration and exclusivity: A cohort study at a Brazilian referral center for high-risk neonates and infants. PLoS One 2021; 16:e0255190. [PMID: 34358227 PMCID: PMC8346259 DOI: 10.1371/journal.pone.0255190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background and aim Both breastfeeding and the use of human milk are strategies that provide better conformation to health throughout an individual’s life and bring countless short- and long- term benefits, which are well established in the scientific literature. For at-risk newborns (NBs), these strategies are crucial interventions to enable neonatal survival with better quality of life due to the distinctive and complex composition of human milk, which serves as personalized food-medicine-protection. However, there is limited knowledge about breastfeeding practices in high-risk NBs. The aim was to estimate the duration of EBF and to investigate the effect of risk at birth on EBF discontinuity in the first six months of life’. Methods This cohort study included 1,003 NBs from a high-risk referral center, followed up from birth to the sixth month of life, between 2017 and 2018. Correspondence and cluster analysis was used to identify neonatal risk clusters as the main exposure. The object of interest was the time until EBF discontinuity. The Kaplan-Meier methods and the Cox proportional hazards model were used to estimate the hazard ratio and 95% confidence intervals. Results The prevalence and median duration of EBF decreased proportionally in the three groups. The multiple model revealed a gradient in EBF discontinuity, which was 40% higher in risk group 1 and 111% higher in risk group 2 compared to healthy full-term NBs. Additionally, EBF during hospitalization predicted a longer median duration of this practice for high-risk NBs. Conclusion This study confirms a high proportion of high-risk NBs who have EBF discontinued before six months of life. The risk of EBF discontinuity is higher in risk groups, with a gradual effect even when adjusted by several factors. Effective interventions are needed to promote, protect, and support breastfeeding in different profiles of risk-at-birth groups.
Collapse
Affiliation(s)
- Maíra Domingues Bernardes Silva
- Human Milk Bank at the National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF) of the Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
- * E-mail:
| | | | | | | |
Collapse
|
33
|
Santiano FE, Campo Verde Arboccó F, Bruna FA, Zyla LE, Sasso CV, Gómez S, Pistone-Creydt V, López-Fontana CM, Carón RW. The epigenetic role of breastfeeding in mammary differentiation. J Dev Orig Health Dis 2021; 12:578-586. [PMID: 33023719 DOI: 10.1017/s2040174420000902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal milk consumption can cause changes in the mammary epithelium of the offspring that result in the expression of molecules involved in the induction of differentiation, reducing the risk of developing mammary cancer later in life. We previously showed that animals that maintained a higher intake of maternal milk had a lower incidence of mammary cancer. In the present study, we evaluated one of the possible mechanisms by which the consumption of maternal milk could modify the susceptibility to mammary carcinogenesis. We used Sprague Dawley rats reared in litters of 3 (L3), 8 (L8), or 12 (L12) pups per mother in order to generate a differential consumption of milk. Whole mounts of mammary glands were performed to analyze the changes in morphology. Using real-time polymerase chain reaction (PCR), we analyzed the expression of mammary Pinc, Tbx3, Stat6, and Gata3 genes. We use the real-time methylation-specific polymerase chain reaction method to assess the methylation status of Stat6 and Gata3 CpG sites. Our findings show an increase in the size of the epithelial tree and a smaller number of ducts called terminal end buds in L3 vs. L12. We observed an increased expression of mRNA of Stat6, Gata3, Tbx3, and a lower expression of Pinc in L3 with respect to L12. Stat6 and Gata3 are more methylated in the CpG islands of the promoter analyzed in L12 vs. L3. In conclusion, the increased consumption of maternal milk during the postnatal stage generates epigenetic and morphological changes associated with the differentiation of the mammary gland.
Collapse
Affiliation(s)
- Flavia E Santiano
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Fiorella Campo Verde Arboccó
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
- Physiology Department, School of Medicine, University of Mendoza, Mendoza, Argentina
| | - Flavia A Bruna
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Leila E Zyla
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
- Physiology Department, School of Medicine, University of Mendoza, Mendoza, Argentina
| | - Corina V Sasso
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Silvina Gómez
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Virginia Pistone-Creydt
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Constanza M López-Fontana
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| | - Rubén W Carón
- Laboratory of Hormones and Cancer Biology, Institute of Medicine and Experimental Biology of Cuyo, IMBECU, CONICET UNCuyo, Mendoza, Argentina
| |
Collapse
|
34
|
Abstract
Human milk is considered the most advantageous source of nourishment for infants. Even though there is no ideal composition of human milk, it still contains a unique combination of components that contribute to brain development. The aim of this review is to provide an overview on the possible correlation of human milk with the neurodevelopment of infants, with a special emphasis on myelination and epigenetic modifications. Research in human milk is a rapidly expanding field and cutting-edge technologies might contribute to identify specific mechanisms underlying the beneficial effects on human milk on neurodevelopment.
Collapse
|
35
|
Meng F, Uniacke-Lowe T, Ryan AC, Kelly AL. The composition and physico-chemical properties of human milk: A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
36
|
Melnik BC, Schmitz G. Pasteurized non-fermented cow's milk but not fermented milk is a promoter of mTORC1-driven aging and increased mortality. Ageing Res Rev 2021; 67:101270. [PMID: 33571703 DOI: 10.1016/j.arr.2021.101270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/16/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Recent epidemiological studies in Sweden, a country with traditionally high milk consumption, revealed that the intake of non-fermented pasteurized milk increased all-cause mortality in a dose-dependent manner. In contrast, the majority of epidemiological and clinical studies report beneficial health effects of fermented milk products, especially of yogurt. It is the intention of this review to delineate potential molecular aging mechanisms related to the intake of non-fermented milk versus yogurt on the basis of mechanistic target of rapamycin complex 1 (mTORC1) signaling. Non-fermented pasteurized milk via its high bioavailability of insulinotropic branched-chain amino acids (BCAAs), abundance of lactose (glucosyl-galactose) and bioactive exosomal microRNAs (miRs) enhances mTORC1 signaling, which shortens lifespan and increases all-cause mortality. In contrast, fermentation-associated lactic acid bacteria metabolize BCAAs and degrade galactose and milk exosomes including their mTORC1-activating microRNAs. The Industrial Revolution, with the introduction of pasteurization and refrigeration of milk, restricted the action of beneficial milk-fermenting bacteria, which degrade milk's BCAAs, galactose and bioactive miRs that synergistically activate mTORC1. This unrecognized behavior change in humans after the Neolithic revolution increased aging-related over-activation of mTORC1 signaling in humans, who persistently consume large quantities of non-fermented pasteurized cow's milk, a potential risk factor for aging and all-cause mortality.
Collapse
|
37
|
Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. Breast Milk: A Source of Functional Compounds with Potential Application in Nutrition and Therapy. Nutrients 2021; 13:1026. [PMID: 33810073 PMCID: PMC8005182 DOI: 10.3390/nu13031026] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is an unbeatable food that covers all the nutritional requirements of an infant in its different stages of growth up to six months after birth. In addition, breastfeeding benefits both maternal and child health. Increasing knowledge has been acquired regarding the composition of breast milk. Epidemiological studies and epigenetics allow us to understand the possible lifelong effects of breastfeeding. In this review we have compiled some of the components with clear functional activity that are present in human milk and the processes through which they promote infant development and maturation as well as modulate immunity. Milk fat globule membrane, proteins, oligosaccharides, growth factors, milk exosomes, or microorganisms are functional components to use in infant formulas, any other food products, nutritional supplements, nutraceuticals, or even for the development of new clinical therapies. The clinical evaluation of these compounds and their commercial exploitation are limited by the difficulty of isolating and producing them on an adequate scale. In this work we focus on the compounds produced using milk components from other species such as bovine, transgenic cattle capable of expressing components of human breast milk or microbial culture engineering.
Collapse
Affiliation(s)
- Cristina Sánchez
- Pharmacy Faculty, San Pablo-CEU University, 28003 Madrid, Spain;
| | - Luis Franco
- Medicine Faculty, Santiago de Compostela University, 15782 Santiago de Compostela, Spain;
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| |
Collapse
|
38
|
Kortesniemi M, Slupsky CM, Aatsinki AK, Sinkkonen J, Karlsson L, Linderborg KM, Yang B, Karlsson H, Kailanto HM. Human milk metabolome is associated with symptoms of maternal psychological distress and milk cortisol. Food Chem 2021; 356:129628. [PMID: 33836356 DOI: 10.1016/j.foodchem.2021.129628] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 12/18/2022]
Abstract
The composition of human milk is subject to considerable variation, but the effects of maternal stress are largely unknown. We studied differences in human milk metabolome between Finnish mothers (n = 120, secretors) with symptoms of prenatal symptoms of psychological distress and milk cortisol concentrations. Human milk samples acquired at 2.5 months postpartum were analyzed using targeted 1H NMR metabolomics. Self-reported scores for depression (EPDS), overall anxiety (SCL-90), and pregnancy-related anxiety (PRAQ) were used to evaluate psychological distress. Prenatal psychological distress was positively associated with concentrations of short-chain fatty acids, caprate, and hypoxanthine (q < 0.0012). Milk cortisol was positively associated with lactate concentration (q < 0.05). Changes in the human milk metabolome were shown to be associated with maternal psychological distress and concentration of milk cortisol in a dissimilarly, suggesting alterations in bacterial and energy metabolism of the mother, respectively.
Collapse
Affiliation(s)
- Maaria Kortesniemi
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland; Department of Nutrition, University of California Davis, Davis, CA, USA.
| | - Carolyn M Slupsky
- Department of Nutrition, University of California Davis, Davis, CA, USA; Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Jari Sinkkonen
- Instrument Centre, Department of Chemistry, University of Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Finland
| | - Kaisa M Linderborg
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland
| | - Baoru Yang
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Finland
| | - Henna-Maria Kailanto
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| |
Collapse
|
39
|
Acevedo N, Alashkar Alhamwe B, Caraballo L, Ding M, Ferrante A, Garn H, Garssen J, Hii CS, Irvine J, Llinás-Caballero K, López JF, Miethe S, Perveen K, Pogge von Strandmann E, Sokolowska M, Potaczek DP, van Esch BCAM. Perinatal and Early-Life Nutrition, Epigenetics, and Allergy. Nutrients 2021; 13:724. [PMID: 33668787 PMCID: PMC7996340 DOI: 10.3390/nu13030724] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Epidemiological studies have shown a dramatic increase in the incidence and the prevalence of allergic diseases over the last several decades. Environmental triggers including risk factors (e.g., pollution), the loss of rural living conditions (e.g., farming conditions), and nutritional status (e.g., maternal, breastfeeding) are considered major contributors to this increase. The influences of these environmental factors are thought to be mediated by epigenetic mechanisms which are heritable, reversible, and biologically relevant biochemical modifications of the chromatin carrying the genetic information without changing the nucleotide sequence of the genome. An important feature characterizing epigenetically-mediated processes is the existence of a time frame where the induced effects are the strongest and therefore most crucial. This period between conception, pregnancy, and the first years of life (e.g., first 1000 days) is considered the optimal time for environmental factors, such as nutrition, to exert their beneficial epigenetic effects. In the current review, we discussed the impact of the exposure to bacteria, viruses, parasites, fungal components, microbiome metabolites, and specific nutritional components (e.g., polyunsaturated fatty acids (PUFA), vitamins, plant- and animal-derived microRNAs, breast milk) on the epigenetic patterns related to allergic manifestations. We gave insight into the epigenetic signature of bioactive milk components and the effects of specific nutrition on neonatal T cell development. Several lines of evidence suggest that atypical metabolic reprogramming induced by extrinsic factors such as allergens, viruses, pollutants, diet, or microbiome might drive cellular metabolic dysfunctions and defective immune responses in allergic disease. Therefore, we described the current knowledge on the relationship between immunometabolism and allergy mediated by epigenetic mechanisms. The knowledge as presented will give insight into epigenetic changes and the potential of maternal and post-natal nutrition on the development of allergic disease.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Bilal Alashkar Alhamwe
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - James Irvine
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kevin Llinás-Caballero
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Juan Felipe López
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
40
|
Nair J, Maheshwari A. Epigenetics in Necrotizing Enterocolitis. Curr Pediatr Rev 2021; 17:172-184. [PMID: 33882811 DOI: 10.2174/1573396317666210421110608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic alterations in our genetic material can lead to heritable changes in the risk, clinical manifestations, course, and outcomes of many diseases. Understanding these epigenetic mechanisms can help in identifying potential therapeutic targets. This is especially important in necrotizing enterocolitis (NEC), where prenatal as well as postnatal factors impact susceptibility to this devastating condition, but our therapeutic options are limited. Developmental factors affecting intestinal structure and function, our immune system, gut microbiome, and postnatal enteral nutrition are all thought to play a prominent role in this disease. In this manuscript, we have reviewed the epigenetic mechanisms involved in NEC. These include key developmental changes in DNA methylation in the immature intestine, the role of long non-coding RNA (lncRNA) in maintaining intestinal barrier function, epigenetic influences of prenatal inflammation on immunological pathways in NEC pathogenesis such as Toll-Like Receptor 4 (TLR4) and epigenetic changes associated with enteral feeding causing upregulation of pro-inflammatory genes. We have assimilated research findings from our own laboratory with an extensive review of the literature utilizing key terms in multiple databases, including PubMed, EMBASE, and Science Direct.
Collapse
Affiliation(s)
- Jayasree Nair
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, MA, United States
| |
Collapse
|
41
|
Emerald B, Kaimala S, Ansari S. Risk factors which influence DNA methylation in childhood obesity. HAMDAN MEDICAL JOURNAL 2021. [DOI: 10.4103/hmj.hmj_15_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
42
|
Mallisetty Y, Mukherjee N, Jiang Y, Chen S, Ewart S, Arshad SH, Holloway JW, Zhang H, Karmaus W. Epigenome-Wide Association of Infant Feeding and Changes in DNA Methylation from Birth to 10 Years. Nutrients 2020; 13:E99. [PMID: 33396735 PMCID: PMC7824231 DOI: 10.3390/nu13010099] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Epigenetic factors have been suggested as mediators of early-life nutrition to future health. Prior studies focused on breastfeeding effects on DNA methylation (DNAm), ignoring other feeding modes. In this analysis of the Isle of Wight birth cohort, feeding modes were categorized as exclusive breastfeeding (EBF), exclusive formula feeding (EFF), and mixed feeding based on whether the respective feeding mode lasted for at least 3 months. In addition, in the past, infant feeding modes were assessed using DNAm at one time point in childhood, not changes of DNAm. In this paper, methylation differences (delta DNAm) were calculated by subtracting residual methylation values at birth from age 10 years (adjusting for cell types and season of blood collection at both ages). These deltas were estimated for all methylation sites where cytosine was followed by guanine (cytosine guanine dinucleotide (CpG) sites). Then, we performed an epigenome-wide association study contrasting EBF, EFF, and mixed feeding with delta DNAm that represents changes in methylation from birth to 10 years. A total of 87 CpGs (EBF: 27 CpGs, EFF: 48 CpGs, mixed: 12 CpGs) were identified using separate linear regression models adjusting for confounders and multiple testing. The sum of all changes in methylation from birth to age 10 years was significantly lower in the EFF group. Correspondingly, the number of CpGs with a methylation decline was 4.7% higher reflecting 13,683 CpGs. Lower methylation related to exclusive formula feeding and its adverse potential for the child's development needs future research to reduce adverse health effects.
Collapse
Affiliation(s)
- Yamini Mallisetty
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Robison Hall, Memphis, TN 38152, USA; (Y.M.); (N.M.); (Y.J.); (H.Z.)
| | - Nandini Mukherjee
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Robison Hall, Memphis, TN 38152, USA; (Y.M.); (N.M.); (Y.J.); (H.Z.)
| | - Yu Jiang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Robison Hall, Memphis, TN 38152, USA; (Y.M.); (N.M.); (Y.J.); (H.Z.)
| | - Su Chen
- Department of Mathematical Science, University of Memphis, Dunn Hall, Memphis, TN 38152, USA;
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - S. Hasan Arshad
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (S.H.A.); (J.W.H.)
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- The David Hide Asthma and Allergy Research Centre, St Mary’s Hospital, Isle of Wight PO30 5TG, UK
| | - John W. Holloway
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK; (S.H.A.); (J.W.H.)
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Robison Hall, Memphis, TN 38152, USA; (Y.M.); (N.M.); (Y.J.); (H.Z.)
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Robison Hall, Memphis, TN 38152, USA; (Y.M.); (N.M.); (Y.J.); (H.Z.)
| |
Collapse
|
43
|
Ongprasert K, Ruangsuriya J, Malasao R, Sapbamrer R, Suppansan P, Ayood P, Kittisakmontri K, Siviroj P. Macronutrient, immunoglobulin a and total antioxidant capacity profiles of human milk from 1 to 24 months: a cross-sectional study in Thailand. Int Breastfeed J 2020; 15:90. [PMID: 33126900 PMCID: PMC7602312 DOI: 10.1186/s13006-020-00333-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 10/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background An extended duration of breastfeeding of up to two years is encouraged by many health authorities, but information regarding the composition of milk after one year postpartum is limited. The goal of this study was to determine the association between the duration of lactation and macronutrient contents, immunoglobulin A (IgA) levels and total antioxidant capacity (TAC) in human milk (HM), from 1 to 24 months postpartum. Methods Cross-sectional milk samples were collected between January and April 2019 from mothers with healthy full-term children who had been lactating for 1 to 24 months. The HM was biochemically analyzed for protein and carbohydrate contents by colorimetric assays. The fat content was determined by capillary centrifugation, and the energy content was calculated from the results of centrifugation assays. IgA levels and TAC were determined by ELISA and a Trolox equivalent antioxidant capacity (TEAC) assay, respectively. Pearson’s correlation coefficient and Spearman’s rank correlation coefficient were used to determine associations between months of lactation and milk composition, and multiple regression analysis was used to assess associations between months of lactation and milk composition adjusted for relevant covariates. Differences were considered significant at p < 0.05. Results One hundred eighty-four milk samples were analyzed. The month of lactation was positively associated with the fat concentration (B = 0.31, SE = 0.09, p = 0.001), energy content (B = 3.11, SE = 0.92, p = 0.001), and IgA (B = 4.17, SE = 1.08, p < 0.001) but negatively associated with the carbohydrate concentration (B = − 0.22, SE = 0.01, p = 0.04). No association was observed between the month of lactation and the protein concentration or TAC after adjustment for maternal age, maternal BMI, birth order, and breastfeeding frequency. Conclusion The duration of lactation was found to be positively associated with the fat, energy, and IgA content in HM for up to two years postpartum, and negatively associated with carbohydrate concentration. More prospective cohort studies are needed to obtain evidence-based knowledge regarding the changes in HM composition throughout the course of lactation.
Collapse
Affiliation(s)
- Krongporn Ongprasert
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Jetsada Ruangsuriya
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Rungnapa Malasao
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Ratana Sapbamrer
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pikul Suppansan
- Maharaj Nakorn Chiang Mai Hospital, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pisittawoot Ayood
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kulnipa Kittisakmontri
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Penprapa Siviroj
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
44
|
Association between Breastfeeding and DNA Methylation over the Life Course: Findings from the Avon Longitudinal Study of Parents and Children (ALSPAC). Nutrients 2020; 12:nu12113309. [PMID: 33137917 PMCID: PMC7692466 DOI: 10.3390/nu12113309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Breastfeeding is associated with short and long-term health benefits. Long-term effects might be mediated by epigenetic mechanisms, yet the literature on this topic is scarce. We performed the first epigenome-wide association study of infant feeding, comparing breastfed vs non-breastfed children. We measured DNA methylation in children from peripheral blood collected in childhood (age 7 years, N = 640) and adolescence (age 15–17 years, N = 709) within the Accessible Resource for Integrated Epigenomic Studies (ARIES) project, part of the larger Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. Cord blood methylation (N = 702) was used as a negative control for potential pre-natal residual confounding. Results: Two differentially-methylated sites presented directionally-consistent associations with breastfeeding at ages 7 and 15–17 years, but not at birth. Twelve differentially-methylated regions in relation to breastfeeding were identified, and for three of them there was evidence of directional concordance between ages 7 and 15–17 years, but not between birth and age 7 years. Conclusions: Our findings indicate that DNA methylation in childhood and adolescence may be predicted by breastfeeding, but further studies with sufficiently large samples for replication are required to identify robust associations.
Collapse
|
45
|
Zeng B, Chen T, Luo JY, Zhang L, Xi QY, Jiang QY, Sun JJ, Zhang YL. Biological Characteristics and Roles of Noncoding RNAs in Milk-Derived Extracellular Vesicles. Adv Nutr 2020; 12:1006-1019. [PMID: 33080010 PMCID: PMC8166544 DOI: 10.1093/advances/nmaa124] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have diverse roles in the transport of proteins, lipids, and nucleic acids between cells, and they serve as mediators of intercellular communication. Noncoding RNAs (ncRNAs) that are present in EVs, including microRNAs, long noncoding RNAs, and circular RNAs, have been found to participate in complex networks of interactions and regulate a wide variety of genes in animals. Milk is an important source of nutrition for humans and other mammals. Evidence suggests that milk-derived EVs contain abundant ncRNAs, which are stable and can be transported to the offspring and other consumers. Current data suggest a strong link between milk EV ncRNAs and many biological processes, and these ncRNAs have been drawing increasing attention and might play an epigenetic regulatory role in recipients, though further research is still necessary to understand their precise roles. The present review introduces basic information about milk EV ncRNAs, summarizes their expression profiles, biological characteristics, and functions based on current knowledge, and discusses their biological roles, indeterminate issues, and perspectives. Our goal is to provide a deeper understanding of the physiological effects of milk EV ncRNAs on offspring and to provide a reference for future research in this field.
Collapse
Affiliation(s)
- Bin Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jun-Yi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qian-Yun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qing-Yan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | | | | |
Collapse
|
46
|
Dhami MV, Ogbo FA, Diallo TM, Agho KE. Regional Analysis of Associations between Infant and Young Child Feeding Practices and Diarrhoea in Indian Children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4740. [PMID: 32630337 PMCID: PMC7370018 DOI: 10.3390/ijerph17134740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 11/07/2022]
Abstract
Studies on the association between infant and young child feeding (IYCF) practices and diarrhoea across regional India are limited. Hence, we examined the association between IYCF practices and diarrhoea in regional India. A weighted sample of 90,596 (North = 11,200, South = 16,469, East = 23,317, West = 11,512, Central = 24,870 and North-East = 3228) from the 2015-2016 National Family Health Survey in India was examined, using multivariate logistic regressions that adjust for clustering and sampling weights. The IYCF indicators included early initiation of breastfeeding (EIBF), exclusive breastfeeding (ExcBF), predominant breastfeeding (PBF), bottle feeding (BotF), continued breastfeeding (BF) at one-year, continued BF at two years, children ever breastfed and the introduction of solid, semi-solid or soft foods (ISSSF). Diarrhoea prevalence was lower among infants who were BF within one-hour of birth and those who were exclusively breastfed. Multivariate analyses revealed that continued BF at one and two years, and infants who were introduced to complementary foods had a higher prevalence of diarrhoea. EIBF and ExcBF were protective against diarrhoea in the regions of North, East and Central India. PBF, BotF and ISSSF were risk factors for diarrhoea in Central India. Continued BF at two years was a risk factor for diarrhoea in Western India. Findings suggested that EIBF and ExcBF were protective against diarrhoea in Northern, Eastern and Central India, while PBF, BotF, continued BF at two years and ISSSF were risk factors for diarrhoea in various regions in India. Improvements in IYCF practices are likely to reduce the burden of diarrhoea-related morbidity and mortality across regions in India.
Collapse
Affiliation(s)
- Mansi Vijaybhai Dhami
- Translational Health Research Institute (THRI), School of Medicine, Western Sydney University, Campbelltown Campus, Penrith, NSW 2571, Australia; (F.A.O.); (K.E.A.)
| | - Felix Akpojene Ogbo
- Translational Health Research Institute (THRI), School of Medicine, Western Sydney University, Campbelltown Campus, Penrith, NSW 2571, Australia; (F.A.O.); (K.E.A.)
- General Practice Unit, Prescot Specialist Medical Centre, Welfare Quarters, Makurdi, Benue State 972261, Nigeria
| | - Thierno M.O. Diallo
- School of Social Sciences, Western Sydney University, Penrith Campus, Penrith, NSW 2571, Australia;
- Statistiques & M. N., Sherbrooke, QC J1K 2Z4, Canada
| | - Kingsley E. Agho
- Translational Health Research Institute (THRI), School of Medicine, Western Sydney University, Campbelltown Campus, Penrith, NSW 2571, Australia; (F.A.O.); (K.E.A.)
- African Vision Research Institute (AVRI), University of KwaZulu-Natal, Durban 4041, South Africa
- School of Health Sciences, Western Sydney University, Campbelltown Campus, Penrith, NSW 2571, Australia
| | | |
Collapse
|
47
|
Harvey SM, Murphy VE, Gibson PG, Collison A, Robinson P, Sly PD, Mattes J, Jensen ME. Maternal asthma, breastfeeding, and respiratory outcomes in the first year of life. Pediatr Pulmonol 2020; 55:1690-1696. [PMID: 32250063 DOI: 10.1002/ppul.24756] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/18/2020] [Indexed: 11/11/2022]
Abstract
Maternal asthma increases the risk of infant wheeze. Breastfeeding may offer protection but there is limited evidence in this high-risk group. We examined associations between breastfeeding and respiratory outcomes, in infants born to women with asthma. This study was a secondary analysis of two prospective cohorts of pregnant women with asthma, and their infants, conducted between 2007 and 2018. At 6 ± 1 (T1) and 12 ± 1 (T2) months post-partum, mothers reported breastfeeding patterns and infant wheeze (primary outcome), bronchiolitis, and related medication use and healthcare utilization, via a validated questionnaire; a subgroup completed face-to-face interviews. χ2 tests and logistic regression models, adjusting for confounders, were utilized. Data were complete for 605 participants at T1 and 486 (80%) at T2. Of 605 participants: 89% initiated breastfeeding and 38% breastfed for more than 6 months. Breastfeeding for more than 6 months vs "never" was associated with a reduced adjusted relative risk of infant wheeze at T1 (0.54, 95% confidence interval, 0.30-0.96). Bronchiolitis risk was reduced at T1 and T2 with more tha 6 months of breastfeeding vs "never." Breastfeeding duration of 1 to 3 months, 4 to 6 months, and more than 6 months were associated with a reduced risk of infant healthcare utilization (all P < .05, vs "never"), but not medication use (P > .05). Breastfeeding for more than 6 months was associated with a reduced risk of wheeze, bronchiolitis, and wheeze-related healthcare utilization in infants at risk due to maternal asthma. Notably, breastfeeding for shorter durations was associated with a reduced risk of healthcare utilization compared with none. Larger cohorts are needed to further examine the impact of breastfeeding exposure on respiratory health in infants exposed to maternal asthma.
Collapse
Affiliation(s)
- Soriah M Harvey
- Respiratory Department, Priority Research Centre Grow Up Well, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Vanessa E Murphy
- Respiratory Department, Priority Research Centre Grow Up Well, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Peter G Gibson
- Respiratory Department, Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Adam Collison
- Respiratory Department, Priority Research Centre Grow Up Well, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Paul Robinson
- Department of Respiratory Medicine, The Childrens Hospital Westmead, Woolcock Institue of Medical Research, Sydney, Australia
| | - Peter D Sly
- Children's Health & Environment, The Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Joerg Mattes
- Respiratory Department, Priority Research Centre Grow Up Well, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Department of Paediatric Respiratory and Sleep Medicine, John Hunter Children's Hospital, Newcastle, Australia
| | - Megan E Jensen
- Respiratory Department, Priority Research Centre Grow Up Well, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| |
Collapse
|
48
|
Sherwood WB, Kothalawala DM, Kadalayil L, Ewart S, Zhang H, Karmaus W, Arshad SH, Holloway JW, Rezwan FI. Epigenome-Wide Association Study Reveals Duration of Breastfeeding Is Associated with Epigenetic Differences in Children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E3569. [PMID: 32443666 PMCID: PMC7277240 DOI: 10.3390/ijerph17103569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/21/2022]
Abstract
Several small studies have shown associations between breastfeeding and genome-wide DNA methylation (DNAm). We performed a comprehensive Epigenome-Wide Association Study (EWAS) to identify associations between breastfeeding and DNAm patterns in childhood. We analysed DNAm data from the Isle of Wight Birth Cohort at birth, 10, 18 and 26 years. The feeding method was categorized as breastfeeding duration >3 months and >6 months, and exclusive breastfeeding duration >3 months. EWASs using robust linear regression were performed to identify differentially methylated positions (DMPs) in breastfed and non-breastfed children at age 10 (false discovery rate of 5%). Differentially methylated regions (DMRs) were identified using comb-p. The persistence of significant associations was evaluated in neonates and individuals at 18 and 26 years. Two DMPs, in genes SNX25 and LINC00840, were significantly associated with breastfeeding duration >6 months at 10 years and was replicated for >3 months of exclusive breastfeeding. Additionally, a significant DMR spanning the gene FDFT1 was identified in 10-year-old children who were exposed to a breastfeeding duration >3 months. None of these signals persisted to 18 or 26 years. This study lends further support for a suggestive role of DNAm in the known benefits of breastfeeding on a child's future health.
Collapse
Affiliation(s)
- William B. Sherwood
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
| | - Dilini M. Kothalawala
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Latha Kadalayil
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, 236A Robison Hall, Memphis, TN 38152, USA; (H.Z.); (W.K.)
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, 236A Robison Hall, Memphis, TN 38152, USA; (H.Z.); (W.K.)
| | - S. Hasan Arshad
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- The David Hide Asthma and Allergy Research Centre, St Mary’s Hospital, Isle of Wight PO30 5TG UK
| | - John W. Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Faisal I. Rezwan
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (W.B.S.); (D.M.K.); (L.K.); (F.I.R.)
- School of Water, Energy and Environment, Cranfield University, Cranfield, Bedfordshire MK43 0AL, UK
| |
Collapse
|
49
|
Trinta VDO, Padilha PDC, Petronilho S, Santelli RE, Braz BF, Freire AS, Saunders C, Rocha HFD, Sanz-Medel A, Fernández-Sánchez ML. Total metal content and chemical speciation analysis of iron, copper, zinc and iodine in human breast milk using high-performance liquid chromatography separation and inductively coupled plasma mass spectrometry detection. Food Chem 2020; 326:126978. [PMID: 32413760 DOI: 10.1016/j.foodchem.2020.126978] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 02/08/2023]
Abstract
The aim of this research was to quantify essential trace elements (iron, copper, zinc and iodine) and establish their speciation in human milk. Both the element and the species are important in new-born nutrition. Colostrum, and transitional and mature milks (25) were collected from 18 mothers of pre-term or full-term infants. Concentrations of the target elements were determined using ICP-MS. For speciation, HPLC coupled to ICP-MS was employed. Total contents of the micronutrients varied in mothers of pre-term (Fe = 0.997, Cu = 0.506, Zn = 4.15 and I = 0.458 mg L-1) and mothers of full-term (Fe = 0.733, Cu = 0.234, Zn = 2.91 and I = 0.255 mg L-1) infants. Fe, Cu and Zn were associated with biomolecules with high molecular mass compounds, such as immunoglobulins, albumin and lactoferrin whilst iodine was only found as iodide.
Collapse
Affiliation(s)
- Vânia de Oliveira Trinta
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil
| | - Patrícia de Carvalho Padilha
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil; Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco J, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Sonaly Petronilho
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil
| | - Ricardo Erthal Santelli
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil.
| | - Bernardo Ferreira Braz
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil
| | - Aline Soares Freire
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil
| | - Cláudia Saunders
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil; Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco J, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Hélio Fernandes da Rocha
- Medical School, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco K, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Alfredo Sanz-Medel
- Physical and Analytical Chemistry Department, Faculty of Chemistry, University of Oviedo, Oviedo, Spain
| | | |
Collapse
|
50
|
Sorce LR, Curley MAQ, Kleinpell R, Swanson B, Meier PP. Mother's Own Milk Feeding and Severity of Respiratory Illness in Acutely Ill Children: An Integrative Review. J Pediatr Nurs 2020; 50:5-13. [PMID: 31670137 DOI: 10.1016/j.pedn.2019.09.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/25/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022]
Abstract
PROBLEM Breastfed infants experience less severe infections while actively breastfeeding. However, little is known about whether a history of prior breastfeeding affects severity of illness. Therefore, the purpose of this integrative review was to examine the relationship between previous exposure to mother's own milk (MOM) feeding and severity of respiratory infectious illness in infants and children. ELIGIBILITY CRITERIA Studies meeting the following criteria were included: human subjects, term birth, ages 0-35 months at time of study, diagnosis of pneumonia, bronchiolitis or croup, MOM feeding, and statistical analyses reporting separate respiratory infectious illness outcomes when combined with other infections. SAMPLE Twelve articles met eligibility criteria. RESULTS Major findings were inconsistent definitions of both dose and exposure period of breastfeeding and the severity of illness. In particular, the severity of illness measure was limited by the use of proxy variables such as emergency room visits or hospitalizations that lacked reliability and validity. However, given this limitation, the data suggested that exclusive breastfeeding for four to six months was associated with reduced severity of illness as measured by frequency of visits to the primary care provider office, emergency department or hospitalization. CONCLUSIONS Future research in this area should incorporate reliable and valid measures of MOM dose and exposure period and severity of illness outcomes in the critically ill child. IMPLICATIONS Among many positive outcomes associated with breastfeeding, an additional talking point for encouragement of exclusive breastfeeding for four to six months may be protective against severe respiratory infectious illness after cessation of breastfeeding.
Collapse
Affiliation(s)
- Lauren R Sorce
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL USA.
| | - Martha A Q Curley
- Ruth M. Colket Endowed Chair in Pediatric Nursing, Children's Hospital of Philadelphia, University of Pennsylvania School of Nursing, Anesthesia and Critical Care Medicine - Perelman School of Medicine, Philadelphia, PA USA.
| | - Ruth Kleinpell
- Rush University College of Nursing, Nashville, TN USA; Vanderbilt University School of Nursing, Nashville, TN USA.
| | - Barbara Swanson
- Adult Health & Gerontological Nursing, Nursing Science Studies, Journal of the Association of Nurses in AIDS Care, Rush University College of Nursing, Chicago, IL USA.
| | - Paula P Meier
- Neonatal Intensive Care, Pediatrics, Women, Children and Family Nursing, Rush University Medical Chicago, IL USA.
| |
Collapse
|