1
|
Quifer-Rada P, Aguilar-Camprubí L, Samino S, Amigó N, Soler O, Padró-Arocas A. Metabolomics Approach to Identify Biomarkers of Acute and Subacute Mastitis in Milk Samples: A Pilot Case-Control Study. Metabolites 2024; 14:566. [PMID: 39452946 PMCID: PMC11509265 DOI: 10.3390/metabo14100566] [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: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Background and aims: Mastitis is one of the main complications during breastfeeding and contributes to the cessation of breastfeeding. However, the etiopathogenesis and diagnosis of mastitis are complex and not yet well defined. We aimed to identify metabolic and lipidic changes in human milk during acute and subacute mastitis in order to detect potential biomarkers of mastitis. Methods: We conducted a pilot case-control study including 14 breastfeeding women with acute mastitis, 32 with subacute mastitis symptoms, and 19 without any mastitis symptoms (control). Milk samples were collected and analyzed by proton nuclear magnetic resonance (H-NMR) for metabolomics analysis. To assess the association between the significant metabolites and lipids and the development of acute and subacute mastitis, multi-adjusted logistic regression models were developed. Results: The NMR-based metabolomics approach was able to identify and quantify a total of 40 metabolites in breast milk samples. After adjusting for confounding variables, acute mastitis was significantly associated with acetate (OR 3.9 IC 1.4-10.8), total cholesterol (OR 14 CI 3.2-62), esterified cholesterol (OR 3.3 CI 1.9-5.8), and sphingomyelin (OR 2.6 CI 1.2-5.8). The other metabolites presented weak association (OR < 2.5). Subacute mastitis was significantly associated with glutamine, lysophosphatidylcholine, phosphatidylcholine, plasmalogen, and total polyunsaturated fatty acids, but only cholesterol showed a strong association (OR > 2.5) with an OR of 2.6 (IC 1.1-6.6). Conclusions: Metabolic alteration in breast milk occurs during a process of both acute and subacute mastitis. Acetate, esterified cholesterol, lysophostidylcholine, and polyunsaturated fatty acids increased in both acute and subacute mastitis. However, according to the multi-adjusted regression logistic models, the candidate biomarkers for acute and subacute mastitis are cholesterol, lysophosphatidylcoholine, phosphatidylcholine, plasmalogen, and polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Paola Quifer-Rada
- LactApp Women Health, 08014 Barcelona, Spain; (P.Q.-R.); (L.A.-C.)
- Department of Endocrinology & Nutrition, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, 28029 Madrid, Spain; (S.S.); (N.A.)
| | | | - Sara Samino
- CIBER of Diabetes and Associated Metabolic Diseases, 28029 Madrid, Spain; (S.S.); (N.A.)
- Biosfer Teslab, 43206 Reus, Spain;
| | - Nuria Amigó
- CIBER of Diabetes and Associated Metabolic Diseases, 28029 Madrid, Spain; (S.S.); (N.A.)
- Biosfer Teslab, 43206 Reus, Spain;
- Department of Basic Medical Sciences, University Rovira I Virgili, IISPV, 43204 Reus, Spain
| | | | | |
Collapse
|
2
|
Astono J, Huang YP, Sundekilde UK, Barile D. Human milk oligosaccharide profiles remain unaffected by maternal pre-pregnancy body mass index in an observational study. Front Nutr 2024; 11:1455251. [PMID: 39479194 PMCID: PMC11523534 DOI: 10.3389/fnut.2024.1455251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are important carbohydrates in human milk that infants cannot digest, acting as prebiotics linked to infant health. The risk of childhood obesity increases with maternal obesity, potentially mediated through the gut microbiota affected by the available HMOs. Studies on whether maternal obesity affects HMO abundance, yield conflicting results. This study aimed to investigate the HMO profile and its association with maternal obesity measured by pre-pregnancy body mass index (BMI) and infant anthropometrics. The results were discussed in the context of existing literature. 90 human milk samples were collected at 3 months postpartum from mothers in three BMI-groups: 32 normal weight (BMI: 18.5-24.99 kg/m2), 34 overweight (BMI: 25-30 kg/m2), and 24 obese (BMI > 30 kg/m2). The samples were analyzed using nano liquid chromatography chip quadrupole time-of-flight mass spectrometry yielding 51 HMO structures and isomers. Their peak areas were integrated and normalized to determine relative abundances. Univariate and multivariate analysis showed associations between relative HMO abundance and donors' secretor status and specific infant anthropometric variables, but not with maternal pre-pregnancy BMI. This study does not support the hypothesis that maternal overweight influences the HMO profile and highlights the importance of reporting results despite absence of significant correlations.
Collapse
Affiliation(s)
- Julie Astono
- Department of Food Science, Aarhus University, Aarhus, Denmark
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Yu-Ping Huang
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | | | - Daniela Barile
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
3
|
Muraglia M, Faienza MF, Tardugno R, Clodoveo ML, Matias De la Cruz C, Bermúdez FG, Munizaga MG, Valencia L, Corbo F, Orellana-Manzano A. Breastfeeding: science and knowledge in pediatric obesity prevention. Front Med (Lausanne) 2024; 11:1430395. [PMID: 39399112 PMCID: PMC11466875 DOI: 10.3389/fmed.2024.1430395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
The increasing prevalence of childhood obesity worldwide is a significant concern due to its link to severe health issues in adulthood, such as non-communicable diseases (NCDs). To address this issue, this review evaluates the effectiveness of various preventive measures for childhood obesity, focusing on maternal nutrition and breastfeeding. The study underscores the criticality of the periconceptional period, where the diets of both parents can influence epigenetic modifications that impact the child's metabolic pathways and obesity risks. Breastfeeding is a potent protective mechanism against early-onset obesity, significantly enhancing the infant's metabolic and immune health by modifying DNA methylation and gene expression. Furthermore, the perspective underscores the significance of the Mediterranean diet during the periconceptional period and lactation. This diet can effectively prevent gestational complications and improve breast milk quality, fostering optimal infant development. Recognizing that obesity results from genetic, epigenetic, environmental, and social factors, the paper advocates for a comprehensive, multidisciplinary approach from the earliest stages of life. This approach champions a balanced maternal diet, exclusive breastfeeding, and timely introduction to complementary foods. In conclusion, addressing pediatric obesity requires a multifaceted strategy emphasizing improving prenatal and postnatal nutrition. Further research is necessary to understand the epigenetic mechanisms influenced by nutrition and their long-term effects on children's health. This will help refine interventions that curb the obesity epidemic among future generations.
Collapse
Affiliation(s)
- Marilena Muraglia
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, Bari, Italy
| | - Roberta Tardugno
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Lisa Clodoveo
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari-Aldo Moro, Bari, Italy
| | - Carmen Matias De la Cruz
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Fátima German Bermúdez
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - María Gabriela Munizaga
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Luz Valencia
- Licenciatura en Nutrición y Dietética, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Filomena Corbo
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Andrea Orellana-Manzano
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| |
Collapse
|
4
|
da Silva Rosa Freire S, Padilha M, Lima Ferreira AL, Machado Schincaglia R, Cunha Figueiredo AC, Freitas-Costa NC, Yin X, Brennan L, Kac G. Association between the third trimester maternal serum metabolome and child growth and development through the first year of life. Sci Rep 2024; 14:18360. [PMID: 39112666 PMCID: PMC11306240 DOI: 10.1038/s41598-024-69247-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Evidence suggests that maternal metabolome may be associated with child health outcomes. We analyzed the association between the maternal metabolome between 28-35 gestational weeks and child growth and development during the first year. A prospective cohort of 98 mother-child dyads was followed at birth, 1, 6, and 12 months. Maternal serum samples were collected for targeted LC-MS/MS analysis, which measured 132 metabolites. The child's growth and development were assessed at each time-point. Z-scores were calculated based on WHO growth standards, and the domains of development were assessed using the Ages and Stages Questionnaires (ASQ-3). Multiple linear mixed-effects models were performed and confounders were identified using a Diagram Acyclic Graph. The Benjamini-Hochberg correction was used for multiple comparison adjustments. We found a positive association between lysophosphatidylcholines (14:0; 16:0; 16:1; 17:0; 18:0; 18:1; 18:2; 20:4) with the z-score of weight-for-age, and lysophosphatidylcholines (14:0; 16:0; 16:1; 18:0) and taurine with the z-score of weight-for-length, and lysophosphatidylcholines (14:0; 16:0; 16:1; 17:0; 18:0; 18:1; 18:2; 20:4) and glycine with the z-score of BMI-for-age. The leucine, methionine, tryptophan, and valine were negatively associated with the fine motor skills domain. We observed an association between maternal metabolome and the growth and child's development throughout the first year.
Collapse
Affiliation(s)
- Samary da Silva Rosa Freire
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marina Padilha
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Lorena Lima Ferreira
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Amanda Caroline Cunha Figueiredo
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Nathalia Cristina Freitas-Costa
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Xiaofei Yin
- UCD School of Agriculture and Food Science, Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Lorraine Brennan
- UCD School of Agriculture and Food Science, Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Gilberto Kac
- Department of Social and Applied Nutrition, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Nutritional Epidemiology, Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, J2, Room 29, Cidade Universitária, Rio de Janeiro, RJ, 21941902, Brazil.
| |
Collapse
|
5
|
Roland CB, Seyedhosseini P, Knudsen SDP, Jessen AD, Jensen IKB, Bendix JM, van Hall G, Molsted S, Alomairah SA, Løkkegaard E, Stallknecht B, Clausen TD. Effects of prenatal exercise interventions on maternal body composition: A secondary analysis of the FitMum randomized controlled trial. PLoS One 2024; 19:e0308214. [PMID: 39088510 PMCID: PMC11293652 DOI: 10.1371/journal.pone.0308214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024] Open
Abstract
The main objective of the study was to investigate the effects of prenatal exercise interventions on maternal body composition at 28 weeks gestation and 7-14 days after delivery. We also explored associations between physical activity (PA) per se and body composition. This study presents secondary outcomes of the FitMum randomized controlled trial, which included healthy inactive pregnant women at gestational age ≤ 15+0 weeks. They were randomized to structured supervised exercise training, motivational counselling on PA, or standard care. Maternal body composition was measured by doubly labeled water at 28 weeks gestation (n = 134) and by dual-energy X-ray absorptiometry scan 7-14 days after delivery (n = 117). PA, including moderate-to-vigorous-intensity PA (MVPA), active kilocalories, and steps, were measured continuously from inclusion to delivery by a wrist-worn activity tracker. One hundred fifty pregnant women were included with a median pre-pregnancy body mass index (BMI) of 24.1 (21.6-27.9) kg/m2. We found no differences between groups in fat mass, fat percentage or fat-free mass at 28 weeks gestation or 7-14 days after delivery. Visceral adipose tissue mass and bone mineral density measured 7-14 days after delivery did not differ between groups either. Linear regression analyses adjusted for pre-pregnancy BMI showed that a higher number of daily steps was associated with lower fat mass, fat percentage, and visceral adipose tissue mass at 28 weeks gestation and 7-14 days after delivery. Active kilocalories during pregnancy was positively associated with fat-free mass 7-14 days after delivery. Neither structured supervised exercise training nor motivational counselling on PA during pregnancy affected maternal body composition at 28 weeks gestation or 7-14 days after delivery compared to standard care. Interestingly, when adjusted for pre-pregnancy BMI, higher number of daily steps was associated with lower fat content during pregnancy and after delivery, whereas MVPA and active kilocalories were not. Trial registration: ClinicalTrials.gov; NCT03679130; 20/09/2018.
Collapse
Affiliation(s)
- Caroline Borup Roland
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Parisa Seyedhosseini
- Department of Nuclear Medicine and Clinical Physiology, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Signe de Place Knudsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Anne Dsane Jessen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Ida Karoline Bach Jensen
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Jane M. Bendix
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
- Department of Clinical Research, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
| | - Gerrit van Hall
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Biochemistry, Clinical Metabolomics Core Facility, Rigshospitalet, Copenhagen, Denmark
| | - Stig Molsted
- Department of Clinical Research, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Saud Abdulaziz Alomairah
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Mental Health, John Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Ellen Løkkegaard
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tine D. Clausen
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital–North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Gynecology, Fertility and Obstetrics, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Johnson KE, Hernandez-Alvarado N, Blackstad M, Heisel T, Allert M, Fields DA, Isganaitis E, Jacobs KM, Knights D, Lock EF, Rudolph MC, Gale CA, Schleiss MR, Albert FW, Demerath EW, Blekhman R. Human cytomegalovirus in breast milk is associated with milk composition and the infant gut microbiome and growth. Nat Commun 2024; 15:6216. [PMID: 39043677 PMCID: PMC11266569 DOI: 10.1038/s41467-024-50282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
Human cytomegalovirus (CMV) is a highly prevalent herpesvirus that is often transmitted to the neonate via breast milk. Postnatal CMV transmission can have negative health consequences for preterm and immunocompromised infants, but any effects on healthy term infants are thought to be benign. Furthermore, the impact of CMV on the composition of the hundreds of bioactive factors in human milk has not been tested. Here, we utilize a cohort of exclusively breastfeeding full-term mother-infant pairs to test for differences in the milk transcriptome and metabolome associated with CMV, and the impact of CMV in breast milk on the infant gut microbiome and infant growth. We find upregulation of the indoleamine 2,3-dioxygenase (IDO) tryptophan-to-kynurenine metabolic pathway in CMV+ milk samples, and that CMV+ milk is associated with decreased Bifidobacterium in the infant gut. Our data indicate two opposing CMV-associated effects on infant growth; with kynurenine positively correlated, and CMV viral load negatively correlated, with infant weight-for-length at 1 month of age. These results suggest CMV transmission, CMV-related changes in milk composition, or both may be modulators of full-term infant development.
Collapse
Affiliation(s)
- Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA.
| | | | - Mark Blackstad
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Timothy Heisel
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mattea Allert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - David A Fields
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Katherine M Jacobs
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal-Fetal Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Dan Knights
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, Department of Biochemistry and Physiology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Cheryl A Gale
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mark R Schleiss
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
7
|
Urrutia-Baca VH, Gutiérrez-Uribe JA, Ramos-Parra PA, Domínguez-Uscanga A, Rodriguez-Gutierrez NA, Chavez-Caraza KL, Martinez-Cano I, Padilla-Garza AS, Ruiz-Villarreal EG, Espiricueta-Candelaria F, Chuck-Hernández C. Exploring the impact of maternal factors and dietary habits on human milk oligosaccharide composition in early breastfeeding among Mexican women. Sci Rep 2024; 14:14685. [PMID: 38918476 PMCID: PMC11199484 DOI: 10.1038/s41598-024-63787-1] [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: 12/12/2023] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Human milk oligosaccharides (HMOs) promote adequate intestinal microbiota development and favor the immune system's maturation and cognitive development. In addition to non-modifiable factors, HMOs composition can be influenced by other factors like body mass index and eating habits, but the reports are discrepant. The aim of this work was to describe the correlation between maternal factors and HMOs concentration in colostrum in 70 women from northeastern Mexico categorized into women with normal weight and women with overweight or obesity. The absolute concentration of six HMOs were significantly lower in women with overweight or obesity compared to women with normal weight (LNFPI p = 0.0021, 2'-FL p = 0.0304, LNT p = 0.0492, LNnT p = 0.00026, 3'-SL p = 0.0476, 6'-SL p = 0.00041). Another main finding was that the frequency of consumption of food groups such as vegetables, fruits and meats was positively correlated to specific HMOs (Poblano chili and 2'-FL; rs = 0.702, p = 0.0012; Orange or tangerine and 3-FL; rs = 0.428, p = 0.0022; Chicken and 2'-FL; rs = 0.615, p = 0.0039). This study contributes to the elucidation of how maternal factors influence the composition of HMOs and opens possibilities for future research aimed at mitigating overweight or obesity, consequently improving the quality of human milk.
Collapse
Affiliation(s)
- Víctor H Urrutia-Baca
- Tecnologico de Monterrey, Institute for Obesity Research, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, NL, Mexico
| | - Janet A Gutiérrez-Uribe
- Tecnologico de Monterrey, Institute for Obesity Research, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, NL, Mexico
| | - Perla A Ramos-Parra
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, NL, Mexico
| | - Astrid Domínguez-Uscanga
- Tecnologico de Monterrey, Institute for Obesity Research, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, NL, Mexico
| | - Nora A Rodriguez-Gutierrez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Ignacio Morones Prieto 3000, 64710, Monterrey, NL, Mexico
| | - Karla L Chavez-Caraza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Ignacio Morones Prieto 3000, 64710, Monterrey, NL, Mexico
| | - Ilen Martinez-Cano
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Ave. Dr. José Eleuterio González 235, 64460, Monterrey, NL, Mexico
| | - Alicia S Padilla-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Ignacio Morones Prieto 3000, 64710, Monterrey, NL, Mexico
| | - Elias G Ruiz-Villarreal
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Ave. Dr. José Eleuterio González 235, 64460, Monterrey, NL, Mexico
| | | | - Cristina Chuck-Hernández
- Tecnologico de Monterrey, Institute for Obesity Research, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, NL, Mexico.
| |
Collapse
|
8
|
Vélez-Ixta JM, Juárez-Castelán CJ, Ramírez-Sánchez D, Lázaro-Pérez NDS, Castro-Arellano JJ, Romero-Maldonado S, Rico-Arzate E, Hoyo-Vadillo C, Salgado-Mancilla M, Gómez-Cruz CY, Krishnakumar A, Piña-Escobedo A, Benitez-Guerrero T, Pizano-Zárate ML, Cruz-Narváez Y, García-Mena J. Post Natal Microbial and Metabolite Transmission: The Path from Mother to Infant. Nutrients 2024; 16:1990. [PMID: 38999737 PMCID: PMC11243545 DOI: 10.3390/nu16131990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The entero-mammary pathway is a specialized route that selectively translocates bacteria to the newborn's gut, playing a crucial role in neonatal development. Previous studies report shared bacterial and archaeal taxa between human milk and neonatal intestine. However, the functional implications for neonatal development are not fully understood due to limited evidence. This study aimed to identify and characterize the microbiota and metabolome of human milk, mother, and infant stool samples using high-throughput DNA sequencing and FT-ICR MS methodology at delivery and 4 months post-partum. Twenty-one mothers and twenty-five infants were included in this study. Our results on bacterial composition suggest vertical transmission of bacteria through breastfeeding, with major changes occurring during the first 4 months of life. Metabolite chemical characterization sheds light on the growing complexity of the metabolites. Further data integration and network analysis disclosed the interactions between different bacteria and metabolites in the biological system as well as possible unknown pathways. Our findings suggest a shared bacteriome in breastfed mother-neonate pairs, influenced by maternal lifestyle and delivery conditions, serving as probiotic agents in infants for their healthy development. Also, the presence of food biomarkers in infants suggests their origin from breast milk, implying selective vertical transmission of these features.
Collapse
Affiliation(s)
- Juan Manuel Vélez-Ixta
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Carmen Josefina Juárez-Castelán
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Daniela Ramírez-Sánchez
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Noemí Del Socorro Lázaro-Pérez
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - José Javier Castro-Arellano
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Silvia Romero-Maldonado
- Unidad de Cuidados Intermedios al Recién Nacido, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
| | - Enrique Rico-Arzate
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Carlos Hoyo-Vadillo
- Departamento de Farmacología, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Marisol Salgado-Mancilla
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Carlos Yamel Gómez-Cruz
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Aparna Krishnakumar
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Alberto Piña-Escobedo
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Tizziani Benitez-Guerrero
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - María Luisa Pizano-Zárate
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
- Unidad de Medicina Familiar No. 4, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Yair Cruz-Narváez
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| |
Collapse
|
9
|
Sivalogan K, Liang D, Accardi C, Diaz-Artiga A, Hu X, Mollinedo E, Ramakrishnan U, Teeny SN, Tran V, Clasen TF, Thompson LM, Sinharoy SS. Human Milk Composition Is Associated with Maternal Body Mass Index in a Cross-Sectional, Untargeted Metabolomics Analysis of Human Milk from Guatemalan Mothers. Curr Dev Nutr 2024; 8:102144. [PMID: 38726027 PMCID: PMC11079463 DOI: 10.1016/j.cdnut.2024.102144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024] Open
Abstract
Background Maternal overweight and obesity has been associated with poor lactation performance including delayed lactogenesis and reduced duration. However, the effect on human milk composition is less well understood. Objectives We evaluated the relationship of maternal BMI on the human milk metabolome among Guatemalan mothers. Methods We used data from 75 Guatemalan mothers who participated in the Household Air Pollution Intervention Network trial. Maternal BMI was measured between 9 and <20 weeks of gestation. Milk samples were collected at a single time point using aseptic collection from one breast at 6 mo postpartum and analyzed using high-resolution mass spectrometry. A cross-sectional untargeted high-resolution metabolomics analysis was performed by coupling hydrophilic interaction liquid chromatography (HILIC) and reverse phase C18 chromatography with mass spectrometry. Metabolic features associated with maternal BMI were determined by a metabolome-wide association study (MWAS), adjusting for baseline maternal age, education, and dietary diversity, and perturbations in metabolic pathways were identified by pathway enrichment analysis. Results The mean age of participants at baseline was 23.62 ± 3.81 y, and mean BMI was 24.27 ± 4.22 kg/m2. Of the total metabolic features detected by HILIC column (19,199 features) and by C18 column (11,594 features), BMI was associated with 1026 HILIC and 500 C18 features. Enriched pathways represented amino acid metabolism, galactose metabolism, and xenobiotic metabolic metabolism. However, no significant features were identified after adjusting for multiple comparisons using the Benjamini-Hochberg false discovery rate procedure (FDRBH < 0.2). Conclusions Findings from this untargeted MWAS indicate that maternal BMI is associated with metabolic perturbations of galactose metabolism, xenobiotic metabolism, and xenobiotic metabolism by cytochrome p450 and biosynthesis of amino acid pathways. Significant metabolic pathway alterations detected in human milk were associated with energy metabolism-related pathways including carbohydrate and amino acid metabolism.This trial was registered at clinicaltrials.gov as NCT02944682.
Collapse
Affiliation(s)
- Kasthuri Sivalogan
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Carolyn Accardi
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Anaite Diaz-Artiga
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Xin Hu
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Erick Mollinedo
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Usha Ramakrishnan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
- Department of Environmental Health, College of Public Health, University of Georgia, Athens, GA, United States
| | - Sami Nadeem Teeny
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - ViLinh Tran
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Thomas F Clasen
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Lisa M Thompson
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Sheela S Sinharoy
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
10
|
Aagaard KM, Barkin SL, Burant CF, Carnell S, Demerath E, Donovan SM, Eneli I, Francis LA, Gilbert-Diamond D, Hivert MF, LeBourgeois MK, Loos RJF, Lumeng JC, Miller AL, Okely AD, Osganian SK, Ramirez AG, Trasande L, Van Horn LV, Wake M, Wright RJ, Yanovski SZ. Understanding risk and causal mechanisms for developing obesity in infants and young children: A National Institutes of Health workshop. Obes Rev 2024; 25:e13690. [PMID: 38204366 DOI: 10.1111/obr.13690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 10/02/2023] [Accepted: 11/21/2023] [Indexed: 01/12/2024]
Abstract
Obesity in children remains a major public health problem, with the current prevalence in youth ages 2-19 years estimated to be 19.7%. Despite progress in identifying risk factors, current models do not accurately predict development of obesity in early childhood. There is also substantial individual variability in response to a given intervention that is not well understood. On April 29-30, 2021, the National Institutes of Health convened a virtual workshop on "Understanding Risk and Causal Mechanisms for Developing Obesity in Infants and Young Children." The workshop brought together scientists from diverse disciplines to discuss (1) what is known regarding epidemiology and underlying biological and behavioral mechanisms for rapid weight gain and development of obesity and (2) what new approaches can improve risk prediction and gain novel insights into causes of obesity in early life. Participants identified gaps and opportunities for future research to advance understanding of risk and underlying mechanisms for development of obesity in early life. It was emphasized that future studies will require multi-disciplinary efforts across basic, behavioral, and clinical sciences. An exposome framework is needed to elucidate how behavioral, biological, and environmental risk factors interact. Use of novel statistical methods may provide greater insights into causal mechanisms.
Collapse
Affiliation(s)
- Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Shari L Barkin
- Department of Pediatrics, Children's Hospital of Richmond, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ellen Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sharon M Donovan
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Illinois, USA
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Illinois, USA
| | - Ihuoma Eneli
- Center for Healthy Weight and Nutrition, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
- Center of Nutrition, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Lori A Francis
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Diane Gilbert-Diamond
- Department of Epidemiology, Medicine and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Monique K LeBourgeois
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ruth J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Julie C Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alison L Miller
- Department of Health Behavior and Health Education, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Anthony D Okely
- School of Health and Society, Faculty of Arts, Social Sciences and Humanities, University of Wollongong, Wollongong, New South Wales, Australia
- llawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
- Department of Sport, Food, and Natural Sciences, Western Norway University of Applied Sciences, Sogndal, Norway
| | - Stavroula K Osganian
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amelie G Ramirez
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Leonardo Trasande
- Department of Pediatrics, New York University (NYU) School of Medicine, New York, New York, USA
- Department of Environmental Medicine, New York University (NYU) School of Medicine, New York, New York, USA
- Department of Population Health, New York University (NYU) School of Medicine, New York, New York, USA
| | - Linda V Van Horn
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois, USA
| | - Melissa Wake
- Population Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| | - Susan Z Yanovski
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
11
|
Pinckard KM, Félix-Soriano E, Hamilton S, Terentyeva R, Baer LA, Wright KR, Nassal D, Esteves JV, Abay E, Shettigar VK, Ziolo MT, Hund TJ, Wold LE, Terentyev D, Stanford KI. Maternal exercise preserves offspring cardiovascular health via oxidative regulation of the ryanodine receptor. Mol Metab 2024; 82:101914. [PMID: 38479548 PMCID: PMC10965826 DOI: 10.1016/j.molmet.2024.101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
OBJECTIVE The intrauterine environment during pregnancy is a critical factor in the development of obesity, diabetes, and cardiovascular disease in offspring. Maternal exercise prevents the detrimental effects of a maternal high fat diet on the metabolic health in adult offspring, but the effects of maternal exercise on offspring cardiovascular health have not been thoroughly investigated. METHODS To determine the effects of maternal exercise on offspring cardiovascular health, female mice were fed a chow (C; 21% kcal from fat) or high-fat (H; 60% kcal from fat) diet and further subdivided into sedentary (CS, HS) or wheel exercised (CW, HW) prior to pregnancy and throughout gestation. Offspring were maintained in a sedentary state and chow-fed throughout 52 weeks of age and subjected to serial echocardiography and cardiomyocyte isolation for functional and mechanistic studies. RESULTS High-fat fed sedentary dams (HS) produced female offspring with reduced ejection fraction (EF) compared to offspring from chow-fed dams (CS), but EF was preserved in offspring from high-fat fed exercised dams (HW) throughout 52 weeks of age. Cardiomyocytes from HW female offspring had increased kinetics, calcium cycling, and respiration compared to CS and HS offspring. HS offspring had increased oxidation of the RyR2 in cardiomyocytes coupled with increased baseline sarcomere length, resulting in RyR2 overactivity, which was negated in female HW offspring. CONCLUSIONS These data suggest a role for maternal exercise to protect against the detrimental effects of a maternal high-fat diet on female offspring cardiac health. Maternal exercise improved female offspring cardiomyocyte contraction, calcium cycling, respiration, RyR2 oxidation, and RyR2 activity. These data present an important, translatable role for maternal exercise to preserve cardiac health of female offspring and provide insight on mechanisms to prevent the transmission of cardiovascular diseases to subsequent generations.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Radmila Terentyeva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Katherine R Wright
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Drew Nassal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, OH, USA
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
12
|
Dinleyici M, Pérez-Brocal V, Arslanoglu S, Aydemir O, Sevuk Ozumut S, Tekin N, Vandenplas Y, Moya A, Dinleyici EC. Composition of Microbiota in Transient and Mature Human Milk: Significant Changes in Large for Gestational Age Group. Nutrients 2024; 16:208. [PMID: 38257101 PMCID: PMC10818272 DOI: 10.3390/nu16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/03/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
The composition of the human milk (HM) microbiota and, consequently, the microorganisms that are passed on to the infant through breastfeeding, can be influenced by various factors such as the mother's health and diet, gestational age, delivery mode, lactation stage, method of infant feeding, and geographical location. The aim of the Human Milk-Gest Study was to compare the microbiota of transient (postpartum 7-15 days) and mature HM (postpartum 45-90 days) of 44 mothers, and to investigate any potential changes associated with preterm birth, mode of delivery, and birth weight in relation to gestational age. The data were classified into five study groups: normal spontaneous delivery-term (NS-T) newborns, cesarean delivery-term (CS-T) newborns, preterm (PT) newborns (with a gestational age of less than 37 weeks), small for gestational age (SGA) newborns, and large for gestational age (LGA) newborns. An analysis of differential abundance was conducted using ANCOM-BC to compare the microbial genera between transient and mature HM samples as well as between other study groups. A significant difference was detected between HM samples at different sampling times and between the study groups (p < 0.01). In transient HM samples, Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group compared to the NS-T, CS-T, PT, and SGA groups. In mature HM samples, Burkholderiaceae_uc, Ralstonia, Pelomonas, and Klebsiella were significantly dominant in the LGA group compared to the NS-T, CS-T, and PT groups, while Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group compared to the SGA group. Differences were also detected between the transient and mature HM samples in the CS-T, PT, SGA, and LGA groups, but no differences occurred in the NS-T groups. In conclusion, we showed that Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group in transient HM and continued in mature HM. The body mass index (BMI) of the mothers in the LGA group was not >30 at conception, however, the maternal BMI at birth and maternal weight gain during pregnancy were higher than in the other groups. The nutritional composition of HM is specifically designed to meet infant nutritional requirements during early life. Evaluating the effects of HM microbiota on infant microbiota composition and short- and long-term health effects in larger studies would be useful.
Collapse
Affiliation(s)
- Meltem Dinleyici
- Department of Social Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26480 Eskisehir, Türkiye;
| | - Vicente Pérez-Brocal
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain; (V.P.-B.)
- CIBER in Epidemiology and Public Health (CIBEResp), 28029 Madrid, Spain
| | - Sertac Arslanoglu
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Medeniyet University, 34720 Istanbul, Türkiye
| | - Ozge Aydemir
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye; (O.A.); (N.T.)
| | - Sibel Sevuk Ozumut
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Medeniyet University, 34720 Istanbul, Türkiye
| | - Neslihan Tekin
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye; (O.A.); (N.T.)
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Unversiteit Brussel, 1090 Brussels, Belgium
| | - Andrés Moya
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain; (V.P.-B.)
- CIBER in Epidemiology and Public Health (CIBEResp), 28029 Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC-UVEG), 46980 Valencia, Spain
| | - Ener Cagri Dinleyici
- Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye
| |
Collapse
|
13
|
de Souza HMR, Pereira TTP, de Sá HC, Alves MA, Garrett R, Canuto GAB. Critical Factors in Sample Collection and Preparation for Clinical Metabolomics of Underexplored Biological Specimens. Metabolites 2024; 14:36. [PMID: 38248839 PMCID: PMC10819689 DOI: 10.3390/metabo14010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
This review article compiles critical pre-analytical factors for sample collection and extraction of eight uncommon or underexplored biological specimens (human breast milk, ocular fluids, sebum, seminal plasma, sweat, hair, saliva, and cerebrospinal fluid) under the perspective of clinical metabolomics. These samples are interesting for metabolomics studies as they reflect the status of living organisms and can be applied for diagnostic purposes and biomarker discovery. Pre-collection and collection procedures are critical, requiring protocols to be standardized to avoid contamination and bias. Such procedures must consider cleaning the collection area, sample stimulation, diet, and food and drug intake, among other factors that impact the lack of homogeneity of the sample group. Precipitation of proteins and removal of salts and cell debris are the most used sample preparation procedures. This review intends to provide a global view of the practical aspects that most impact results, serving as a starting point for the designing of metabolomic experiments.
Collapse
Affiliation(s)
- Hygor M. R. de Souza
- Instituto de Química, Universidade Federal do Rio de Janeiro, LabMeta—LADETEC, Rio de Janeiro 21941-598, Brazil;
| | - Tássia T. P. Pereira
- Departamento de Genética, Ecologia e Evolucao, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- Departamento de Biodiversidade, Evolução e Meio Ambiente, Universidade Federal de Ouro Preto, Ouro Preto 35400-000, Brazil
| | - Hanna C. de Sá
- Departamento de Química Analítica, Instituto de Química, Universidade Federal da Bahia, Salvador 40170-115, Brazil;
| | - Marina A. Alves
- Instituto de Pesquisa de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-599, Brazil;
| | - Rafael Garrett
- Instituto de Química, Universidade Federal do Rio de Janeiro, LabMeta—LADETEC, Rio de Janeiro 21941-598, Brazil;
- Department of Laboratory Medicine, Boston Children’s Hospital—Harvard Medical School, Boston, MA 02115, USA
| | - Gisele A. B. Canuto
- Departamento de Química Analítica, Instituto de Química, Universidade Federal da Bahia, Salvador 40170-115, Brazil;
| |
Collapse
|
14
|
Schönbacher L, Treichler C, Brandl W, Köfeler HC, Fluhr H, Jantscher‐Krenn E, van Poppel MNM. Prenatal Human Milk Oligosaccharides (HMOs) in the Context of BMI, Gestational Weight Gain, and Lipid Profile-An Association Study in Pregnant Women with Overweight or Obesity. Mol Nutr Food Res 2024; 68:e2300533. [PMID: 38085123 PMCID: PMC10909570 DOI: 10.1002/mnfr.202300533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/26/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Human milk oligosaccharides (HMOs) are bioactive glycans first detected in human milk. Their presence in maternal blood during pregnancy suggests systemic functions. Dynamics and associations of the most abundant prenatal HMOs in relation to maternal BMI and serum lipids in a cohort of 87 pregnant women with either overweight or obesity are studied. METHODS Serum HMOs (2'FL, 3'SL, 3'SLN, LDFT), serum lipids (total cholesterol, HDL, LDL, triglycerides), and BMI are measured at 15, 24, and 32 weeks of gestation. RESULTS 2'FL and LDFT are negatively correlated to pre-pregnancy BMI and increase significantly slower between 15 and 24 weeks in highly obese women. Women without detectable increase of serum 2'FL (non-secretors) show a less pronounced gestational weight gain and lower BMI in the third trimester as compared to women phenotype as secretors. Higher early-pregnancy 2'FL is associated with high HDL and low triglycerides in pregnancy. On the other hand, higher 3'SL at 15 weeks is associated with higher triglycerides, LDL, and total cholesterol. CONCLUSIONS Higher early-pregnancy 2'FL is associated with a cardioprotective lipid profile, whereas higher 3'SL is associated with an atherogenic lipid profile. Serum trajectories of 2'FL and LDFT in obese women suggest an obesity mediated delay of α-1,2-fucosylation.
Collapse
Affiliation(s)
- Lukas Schönbacher
- Department of Obstetrics and GynecologyMedical University of GrazAuenbruggerplatz 14Graz8036Austria
| | - Carmen Treichler
- Department of Obstetrics and GynecologyMedical University of GrazAuenbruggerplatz 14Graz8036Austria
| | - Waltraud Brandl
- Department of Obstetrics and GynecologyMedical University of GrazAuenbruggerplatz 14Graz8036Austria
| | - Harald C. Köfeler
- BioTechMed‐GrazMozartgasse 12/IIGraz8010Austria
- Center for Medical ResearchMedical University of GrazStiftingtalstraße 24Graz8010Austria
| | - Herbert Fluhr
- Department of Obstetrics and GynecologyMedical University of GrazAuenbruggerplatz 14Graz8036Austria
| | - Evelyn Jantscher‐Krenn
- Department of Obstetrics and GynecologyMedical University of GrazAuenbruggerplatz 14Graz8036Austria
- BioTechMed‐GrazMozartgasse 12/IIGraz8010Austria
| | - Mireille N. M. van Poppel
- BioTechMed‐GrazMozartgasse 12/IIGraz8010Austria
- Institute of Human Movement ScienceSport and HealthUniversity of GrazMozartgasse 14/IGraz8010Austria
| |
Collapse
|
15
|
Moholdt T, Stanford KI. Exercised breastmilk: a kick-start to prevent childhood obesity? Trends Endocrinol Metab 2024; 35:23-30. [PMID: 37735048 PMCID: PMC11005327 DOI: 10.1016/j.tem.2023.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023]
Abstract
Exercise has systemic health benefits through effects on multiple tissues, with intertissue communication. Recent studies indicate that exercise may improve breastmilk composition and thereby reduce the intergenerational transmission of obesity. Even if breastmilk is considered optimal infant nutrition, there is evidence for variations in its composition between mothers who are normal weight, those with obesity, and those who are physically active. Nutrition early in life is important for later-life susceptibility to obesity and other metabolic diseases, and maternal exercise may provide protection against the development of metabolic disease. Here we summarize recent research on the influence of maternal obesity on breastmilk composition and discuss the potential role of exercise-induced adaptations to breastmilk as a kick-start to prevent childhood obesity.
Collapse
Affiliation(s)
- Trine Moholdt
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway.
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
16
|
Brockway MM, Daniel AI, Reyes SM, Granger M, McDermid JM, Chan D, Refvik R, Sidhu KK, Musse S, Patel PP, Monnin C, Lotoski L, Geddes D, Jehan F, Kolsteren P, Allen LH, Hampel D, Eriksen KG, Rodriguez N, Azad MB. Human Milk Macronutrients and Child Growth and Body Composition in the First Two Years: A Systematic Review. Adv Nutr 2024; 15:100149. [PMID: 37981047 PMCID: PMC10831902 DOI: 10.1016/j.advnut.2023.100149] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023] Open
Abstract
Among exclusively breastfed infants, human milk (HM) provides complete nutrition in the first mo of life and remains an important energy source as long as breastfeeding continues. Consisting of digestible carbohydrates, proteins, and amino acids, as well as fats and fatty acids, macronutrients in human milk have been well studied; however, many aspects related to their relationship to growth in early life are still not well understood. We systematically searched Medline, EMBASE, the Cochrane Library, Scopus, and Web of Science to synthesize evidence published between 1980 and 2022 on HM components and anthropometry through 2 y of age among term-born healthy infants. From 9992 abstracts screened, 57 articles reporting observations from 5979 dyads were included and categorized based on their reporting of HM macronutrients and infant growth. There was substantial heterogeneity in anthropometric outcome measurement, milk collection timelines, and HM sampling strategies; thus, meta-analysis was not possible. In general, digestible carbohydrates were positively associated with infant weight outcomes. Protein was positively associated with infant length, but no associations were reported for infant weight. Finally, HM fat was not consistently associated with any infant growth metrics, though various associations were reported in single studies. Fatty acid intakes were generally positively associated with head circumference, except for docosahexaenoic acid. Our synthesis of the literature was limited by differences in milk collection strategies, heterogeneity in anthropometric outcomes and analytical methodologies, and by insufficient reporting of results. Moving forward, HM researchers should accurately record and account for breastfeeding exclusivity, use consistent sampling protocols that account for the temporal variation in HM macronutrients, and use reliable, sensitive, and accurate techniques for HM macronutrient analysis.
Collapse
Affiliation(s)
- Meredith Merilee Brockway
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Canada; Department of Pediatrics and Child Health, University of Manitoba, Canada; Faculty of Nursing, University of Calgary, Canada
| | - Allison I Daniel
- Centre for Global Child Health, Hospital for Sick Children, Canada; Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Sarah M Reyes
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Canada
| | - Matthew Granger
- Department of Food and Human Nutritional Sciences, University of Manitoba, Canada
| | | | - Deborah Chan
- Department of Pediatrics and Child Health, University of Manitoba, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Canada
| | - Rebecca Refvik
- Department of Food and Human Nutritional Sciences, University of Manitoba, Canada
| | - Karanbir K Sidhu
- Department of Food and Human Nutritional Sciences, University of Manitoba, Canada
| | - Suad Musse
- Department of Food and Human Nutritional Sciences, University of Manitoba, Canada
| | - Pooja P Patel
- Department of Public Health and Community Medicine, Tufts University School of Medicine, USA
| | - Caroline Monnin
- Neil John Maclean Health Sciences Library, University of Manitoba, Canada
| | - Larisa Lotoski
- Department of Pediatrics and Child Health, University of Manitoba, Canada
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Australia
| | - Fyezah Jehan
- Department of Pediatrics, Aga Khan University, Pakistan
| | - Patrick Kolsteren
- Department of Food Safety and Food Quality, Ghent University, Belgium
| | - Lindsay H Allen
- Western Human Nutrition Research Center, Agriculture Research Service, United States Department of Agriculture, USA; Department of Nutrition, University of California, Davis, USA
| | - Daniela Hampel
- Western Human Nutrition Research Center, Agriculture Research Service, United States Department of Agriculture, USA; Department of Nutrition, University of California, Davis, USA
| | - Kamilla G Eriksen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Natalie Rodriguez
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Canada; Department of Pediatrics and Child Health, University of Manitoba, Canada
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Canada; Department of Pediatrics and Child Health, University of Manitoba, Canada.
| |
Collapse
|
17
|
Brockway MM, Daniel AI, Reyes SM, Gauglitz JM, Granger M, McDermid JM, Chan D, Refvik R, Sidhu KK, Musse S, Patel PP, Monnin C, Lotoski L, Geddes DT, Jehan F, Kolsteren P, Bode L, Eriksen KG, Allen LH, Hampel D, Rodriguez N, Azad MB. Human Milk Bioactive Components and Child Growth and Body Composition in the First 2 Years: A Systematic Review. Adv Nutr 2024; 15:100127. [PMID: 37802214 PMCID: PMC10831900 DOI: 10.1016/j.advnut.2023.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
Human milk (HM) contains macronutrients, micronutrients, and a multitude of other bioactive factors, which can have a long-term impact on infant growth and development. We systematically searched MEDLINE, EMBASE, Cochrane Library, Scopus, and Web of Science to synthesize evidence published between 1980 and 2022 on HM components and anthropometry through 2 y of age among term-born infants. From 9992 abstracts screened, 141 articles were included and categorized based on their reporting of HM micronutrients, macronutrients, or bioactive components. Bioactives including hormones, HM oligosaccharides (HMOs), and immunomodulatory components are reported here, based on 75 articles from 69 unique studies reporting observations from 9980 dyads. Research designs, milk collection strategies, sampling times, geographic and socioeconomic settings, reporting practices, and outcomes varied considerably. Meta-analyses were not possible because data collection times and reporting were inconsistent among the studies included. Few measured infant HM intake, adjusted for confounders, precisely captured breastfeeding exclusivity, or adequately described HM collection protocols. Only 5 studies (6%) had high overall quality scores. Hormones were the most extensively examined bioactive with 46 articles (n = 6773 dyads), compared with 13 (n = 2640 dyads) for HMOs and 12 (n = 1422 dyads) for immunomodulatory components. Two studies conducted untargeted metabolomics. Leptin and adiponectin demonstrated inverse associations with infant growth, although several studies found no associations. No consistent associations were found between individual HMOs and infant growth outcomes. Among immunomodulatory components in HM, IL-6 demonstrated inverse relationships with infant growth. Current research on HM bioactives is largely inconclusive and is insufficient to address the complex composition of HM. Future research should ideally capture HM intake, use biologically relevant anthropometrics, and integrate components across categories, embracing a systems biology approach to better understand how HM components work independently and synergistically to influence infant growth.
Collapse
Affiliation(s)
- Meredith Merilee Brockway
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Faculty of Nursing, University of Calgary, Calgary, AB, Canada
| | - Allison I Daniel
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Sarah M Reyes
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | | | - Matthew Granger
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | - Deborah Chan
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montréal, QC, Canada
| | - Rebecca Refvik
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Karanbir K Sidhu
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Suad Musse
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Pooja P Patel
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, Unites States
| | - Caroline Monnin
- Neil John Maclean Health Sciences Library, University of Manitoba, Winnipeg, MB, Canada
| | - Larisa Lotoski
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Fyezah Jehan
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan
| | - Patrick Kolsteren
- Department of Food Safety and Food Quality, Ghent University, Ghent, Belgium
| | - Lars Bode
- Department of Pediatrics, Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego (UC San Diego), San Diego, CA, United States
| | - Kamilla G Eriksen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Lindsay H Allen
- Department of Nutrition, University of California, Davis, CA, United States; Western Human Nutrition Research Center, Agriculture Research Service, United States Department of Agriculture, Washington, DC, Unites States
| | - Daniela Hampel
- Department of Nutrition, University of California, Davis, CA, United States; Western Human Nutrition Research Center, Agriculture Research Service, United States Department of Agriculture, Washington, DC, Unites States
| | - Natalie Rodriguez
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
18
|
Holmen M, Giskeødegård GF, Moholdt T. High-intensity exercise increases breast milk adiponectin concentrations: a randomised cross-over study. Front Nutr 2023; 10:1275508. [PMID: 38164413 PMCID: PMC10757973 DOI: 10.3389/fnut.2023.1275508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Adiponectin plays a role in glucose and fat metabolism and is present in human breast milk. It has been postulated that higher breast milk adiponectin concentrations may prevent rapid weight gain in infancy. Prior research indicates that circulating adiponectin increases acutely after endurance exercise, but no prior research has investigated the effect of exercise on breast milk adiponectin concentrations. The purpose of this randomised, cross-over study was to determine the acute effects of endurance exercise on adiponectin concentrations in human breast milk. Methods Participants who were exclusively breastfeeding a 6-12 week-old term infant (N = 20) completed three conditions in the laboratory: (1) Moderate-intensity continuous training (MICT), (2) High-intensity interval training (HIIT), and (3) No activity (REST). At each condition, we collected breast milk at 07:00 h (before exercise/rest), 11:00 h (immediately after exercise/rest), 12:00 h (1 h after exercise/rest), and 15:00 h (4 h after exercise/rest) and determined adiponectin concentrations using enzyme-linked immunosorbent assay. We compared changes in adiponectin concentrations after MICT and HIIT, adjusted for the morning concentration on each test day, with those after REST, using paired t-tests. Results Adiponectin concentrations increased 1 h after HIIT, from 4.6 (± 2.2) μg/L in the 07:00 h sample to 5.6 (± 2.6) μg/L. This change was 0.9 μg/L (95% confidence interval 0.3 to 1.5) greater than the change between these two timepoints in the REST condition (p = 0.025). There were no other statistically significant changes in adiponectin concentrations. Conclusion HIIT may increase adiponectin concentrations in breast milk acutely after exercise. Further studies should determine the impact of exercise-induced elevations in breast milk adiponectin concentrations on growth and metabolism in infancy.
Collapse
Affiliation(s)
- Mads Holmen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Guro F. Giskeødegård
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trine Moholdt
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Women’s Clinic, St.Olavs Hospital, Trondheim, Norway
| |
Collapse
|
19
|
Froń A, Orczyk-Pawiłowicz M. Understanding the Immunological Quality of Breast Milk in Maternal Overweight and Obesity. Nutrients 2023; 15:5016. [PMID: 38140275 PMCID: PMC10746120 DOI: 10.3390/nu15245016] [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: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Maternal obesity, affecting many pregnant women globally, not only poses immediate health risks but also modulates breast milk composition. Obesity is linked to inflammation and oxidative stress, impacting breast milk's immune properties. This paper explores the intricate relationship between maternal metabolic disorders, such as obesity, and breast milk's immunological components. We conducted a thorough search for original and review articles published until 17 October 2023 in the PUBMED/Scopus database. This search included several terms related to human breast milk, immunological properties, and obesity. Articles were selected with the consensus of all authors. Maternal metabolic disorders have discernible effects on the composition of immune-related components in breast milk, such as immunoglobulins, lactoferrin, leptin, ghrelin, adiponectin, C-reactive protein, growth factors, extracellular vesicles, and lymphocytes. These changes in breast milk composition can significantly impact the newborn's immune system, with potential long-term health implications beyond the immediate postnatal period. Maternal metabolic health is a critical factor in shaping the health trajectory of the neonate through breastfeeding, although the full advantages of breastfeeding for children of mothers with obesity remain uncertain. Ongoing research aims to understand and unravel these links.
Collapse
Affiliation(s)
- Anita Froń
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | - Magdalena Orczyk-Pawiłowicz
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
20
|
Moholdt T, Ashby ER, Tømmerdal KH, Lemoine MCC, Holm RL, Sætrom P, Iversen AC, Ravi A, Simpson MR, Giskeødegård GF. Randomised controlled trial of exercise training during lactation on breast milk composition in breastfeeding people with overweight/obesity: a study protocol for the MILKSHAKE trial. BMJ Open Sport Exerc Med 2023; 9:e001751. [PMID: 37829712 PMCID: PMC10565229 DOI: 10.1136/bmjsem-2023-001751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Breast milk from people with overweight/obesity may differ in composition compared with that from normal-weight people. Exercise training can modify breast milk composition in rodent models, with a beneficial impact demonstrated on the offspring's metabolism, but whether these findings translate to humans is unclear. This trial aims to determine the effect of an exercise intervention on breast milk composition and whether an exercise-induced modification of breast milk impacts the infants' growth and body composition. Effect of Exercise Training on Breastmilk Composition is a randomised, controlled trial with two parallel groups, one exercise group and one control group, with a 1:1 allocation. We will include a minimum of 62 exclusively breastfeeding participants, 6 weeks postpartum. The exercise intervention lasts 8 weeks and comprises 25 supervised endurance exercise sessions with moderate or high intensity. The primary outcome measure is the change in the relative concentration of the human milk oligosaccharide 3'sialyllactose in breast milk from baseline at 6 weeks postpartum to the end of the intervention period. Secondary outcomes include breast milk concentrations of other metabolites, cytokines, hormones and microRNA, maternal health outcomes, infant growth, infant gut microbiome and infant circulating microRNA. Maternal and infant outcomes will be measured before, during and after the intervention period, with a follow-up of the infants until they are 24 months old. Trial registration number NCT05488964.
Collapse
Affiliation(s)
- Trine Moholdt
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Gynaecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway
| | - Emily Rose Ashby
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karina Hammer Tømmerdal
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Rebecca Lyng Holm
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Sætrom
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ann-Charlotte Iversen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anuradha Ravi
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Guro F Giskeødegård
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
21
|
Lemas DJ, Du X, Dado-Senn B, Xu K, Dobrowolski A, Magalhães M, Aristizabal-Henao JJ, Young BE, Francois M, Thompson LA, Parker LA, Neu J, Laporta J, Misra BB, Wane I, Samaan S, Garrett TJ. Untargeted Metabolomic Analysis of Lactation-Stage-Matched Human and Bovine Milk Samples at 2 Weeks Postnatal. Nutrients 2023; 15:3768. [PMID: 37686800 PMCID: PMC10490210 DOI: 10.3390/nu15173768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Epidemiological data demonstrate that bovine whole milk is often substituted for human milk during the first 12 months of life and may be associated with adverse infant outcomes. The objective of this study is to interrogate the human and bovine milk metabolome at 2 weeks of life to identify unique metabolites that may impact infant health outcomes. Human milk (n = 10) was collected at 2 weeks postpartum from normal-weight mothers (pre-pregnant BMI < 25 kg/m2) that vaginally delivered term infants and were exclusively breastfeeding their infant for at least 2 months. Similarly, bovine milk (n = 10) was collected 2 weeks postpartum from normal-weight primiparous Holstein dairy cows. Untargeted data were acquired on all milk samples using high-resolution liquid chromatography-high-resolution tandem mass spectrometry (HR LC-MS/MS). MS data pre-processing from feature calling to metabolite annotation was performed using MS-DIAL and MS-FLO. Our results revealed that more than 80% of the milk metabolome is shared between human and bovine milk samples during early lactation. Unbiased analysis of identified metabolites revealed that nearly 80% of milk metabolites may contribute to microbial metabolism and microbe-host interactions. Collectively, these results highlight untargeted metabolomics as a potential strategy to identify unique and shared metabolites in bovine and human milk that may relate to and impact infant health outcomes.
Collapse
Affiliation(s)
- Dominick J. Lemas
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
- Center for Perinatal Outcomes Research, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Xinsong Du
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Bethany Dado-Senn
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Ke Xu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Amanda Dobrowolski
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Marina Magalhães
- Department of Behavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL 32603, USA;
| | - Juan J. Aristizabal-Henao
- Department of Physiological Science, Center for Environmental and Human Toxicology, College of Veterinary Science, University of Florida, Gainesville, FL 32608, USA;
| | - Bridget E. Young
- Division of Breastfeeding and Lactation Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Magda Francois
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Lindsay A. Thompson
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Leslie A. Parker
- Center for Perinatal Outcomes Research, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Josef Neu
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Jimena Laporta
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
| | | | - Ismael Wane
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Samih Samaan
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (X.D.); (K.X.); (A.D.); (M.F.); (L.A.T.); (I.W.); (S.S.)
| | - Timothy J. Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32608, USA;
| |
Collapse
|
22
|
Johnson KE, Heisel T, Fields DA, Isganaitis E, Jacobs KM, Knights D, Lock EF, Rudolph MC, Gale CA, Schleiss MR, Albert FW, Demerath EW, Blekhman R. Human Cytomegalovirus in breast milk is associated with milk composition, the infant gut microbiome, and infant growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549370. [PMID: 37503212 PMCID: PMC10370112 DOI: 10.1101/2023.07.19.549370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Human cytomegalovirus (CMV) is a highly prevalent herpesvirus that is often transmitted to the neonate via breast milk. Postnatal CMV transmission can have negative health consequences for preterm and immunocompromised infants, but any effects on healthy term infants are thought to be benign. Furthermore, the impact of CMV on the composition of the hundreds of bioactive factors in human milk has not been tested. Here, we utilize a cohort of exclusively breastfeeding full term mother-infant pairs to test for differences in the milk transcriptome and metabolome associated with CMV, and the impact of CMV in breast milk on the infant gut microbiome and infant growth. We find upregulation of the indoleamine 2,3- dioxygenase (IDO) tryptophan-to-kynurenine metabolic pathway in CMV+ milk samples, and that CMV+ milk is associated with decreased Bifidobacterium in the infant gut. Our data indicate a complex relationship between milk CMV, milk kynurenine, and infant growth; with kynurenine positively correlated, and CMV viral load negatively correlated, with infant weight-for-length at 1 month of age. These results suggest CMV transmission, CMV-related changes in milk composition, or both may be modulators of full term infant development.
Collapse
Affiliation(s)
- Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Timothy Heisel
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - David A Fields
- Department of Pediatrics, Diabetes-Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Elvira Isganaitis
- Pediatric, Adolescent and Young Adult Unit, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Katherine M Jacobs
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal-Fetal Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Dan Knights
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, Department of Physiology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Cheryl A Gale
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mark R Schleiss
- Division of Pediatric Infectious Diseases and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Londoño-Sierra DC, Mesa V, Guzmán NC, Bolívar Parra L, Montoya-Campuzano OI, Restrepo-Mesa SL. Maternal Diet May Modulate Breast Milk Microbiota-A Case Study in a Group of Colombian Women. Microorganisms 2023; 11:1812. [PMID: 37512984 PMCID: PMC10384792 DOI: 10.3390/microorganisms11071812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
There is increasing evidence that the diet and nutritional status of women during pregnancy and lactation can modulate the microbiota of their milk and, therefore, the microbiota of the infant. An observational, descriptive, and cross-sectional study was carried out in a group of lactating women. Dietary intake during gestation and the first trimester of lactation was evaluated, and the microbiota was analyzed by 16S ribosomal RNA (rRNA) sequencing using the Illumina platform. Globally, Streptococcus spp. (32%), Staphylococcus spp. (17.3%), Corynebacterium spp. (5.1%) and Veillonella spp. (3.1%) were the predominant bacterial genera. The consumption of simple carbohydrates in gestation (rho = 0.55, p ≤ 0.01) and lactation (rho = 0.50, p ≤ 0.01) were positively correlated with Enterobacter spp. In lactation, a negative correlation was observed between the intake of simple carbohydrates and the genus Bifidobacterium spp. (rho = -0.51 p ≤ 0.01); furthermore, a positive correlation was identified between the intake of folic acid and Akkermansia spp. (rho = 0.47, p ≤ 0.01). Amplicon sequence variants (ASVs) associated with the delivery mode, employment relationship, the baby's gender, birth weight, the Body Mass Index (BMI) of the breastfeeding woman, and gestational weight gain were recovered as covariates in a linear mixed model. The results of this research showed that the maternal nutritional status and diet of women during gestation and lactation could modulate the microbiota of breast milk.
Collapse
Affiliation(s)
- Diana C Londoño-Sierra
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Antioquia University, Medellín 050010, Colombia
| | - Victoria Mesa
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Antioquia University, Medellín 050010, Colombia
- Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal (3PHM), INSERM, UMR-S 1139, Université Paris Cité, 75006 Paris, France
| | - Nathalia Correa Guzmán
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Antioquia University, Medellín 050010, Colombia
| | - Laura Bolívar Parra
- Probiotics and Bioprospecting Research Group, Faculty of Sciences, National University of Colombia, Medellín 050034, Colombia
| | - Olga I Montoya-Campuzano
- Probiotics and Bioprospecting Research Group, Faculty of Sciences, National University of Colombia, Medellín 050034, Colombia
| | - Sandra L Restrepo-Mesa
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Antioquia University, Medellín 050010, Colombia
| |
Collapse
|
24
|
Blanco E, Martinez SM, East P, Burrows R, Correa-Burrows P, Lozoff B, Gahagan S. Breastfeeding Duration and Timing of Bottle Supplementation: Associations with Body Mass Index from Childhood to Young-Adulthood. Nutrients 2023; 15:3121. [PMID: 37513539 PMCID: PMC10384694 DOI: 10.3390/nu15143121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Evidence for the association between breastfeeding (BF) duration and later body mass index (BMI) is inconsistent. We explored how BF duration and BF type (exclusive or partial) related to BMI from childhood to young adulthood in a Chilean cohort. Infants were recruited at 6 months between 1994 and 1996 in Santiago, Chile (n = 821). Mothers reported date of first bottle and last BF; anthropometry was measured at 1, 5, 10, 16, and 23 years. We tested whether: (1) type of BF at 6 months (none, partial, exclusive) and (2) duration of exclusive BF (<1 month, 1 to <3 months, 3 to <6 months, and ≥6 months) related to BMI. At 6 months, 35% received both breastmilk and formula ("partial BF") and 38% were exclusively breastfed. We found some evidence of an association between longer BF and lower BMI z-scores at young ages but observed null effects for later BMI. Specifically, BF for 3 to <6 months compared to <1 month related to lower BMI z-scores at 1 and 5 years (both p < 0.05). Our results are in partial accordance with others who have not found a protective effect of longer BF for lower BMI.
Collapse
Affiliation(s)
- Estela Blanco
- Centro de Investigación en Sociedad y Salud y Núcleo Milenio de Sociomedicina, Las Condes, Santiago 7550000, Chile
| | - Suzanna M Martinez
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 90095, USA
| | - Patricia East
- Department of Pediatrics, Division of Child Development and Community Health, University of California, San Diego, CA 92093, USA
| | - Raquel Burrows
- Institute of Nutrition and Food Technology, University of Chile, Macul, Santiago 7810000, Chile
| | - Paulina Correa-Burrows
- Institute of Nutrition and Food Technology, University of Chile, Macul, Santiago 7810000, Chile
| | - Betsy Lozoff
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sheila Gahagan
- Department of Pediatrics, Division of Child Development and Community Health, University of California, San Diego, CA 92093, USA
| |
Collapse
|
25
|
Strobel KM, Juul SE, Hendrixson DT. Maternal Nutritional Status and the Microbiome across the Pregnancy and the Post-Partum Period. Microorganisms 2023; 11:1569. [PMID: 37375071 DOI: 10.3390/microorganisms11061569] [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: 05/15/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Appropriate nutrition during pregnancy and the post-partum period is vital for both the mothers and their offspring. Both under- and over-nourished status may have important microbial implications on the maternal and infant gut microbiomes. Alterations in the microbiome can have implications for a person's risk of obesity and metabolic diseases. In this review, we examine alterations in the maternal gut, vaginal, placental, and milk microbiomes in the context of pre-pregnancy BMI, gestational weight gain, body composition, gestational diabetes, and maternal diet. We also investigate how the infant gut microbiome may be altered by these different parameters. Many of the microbial changes seen in under- and over-nourished states in birthing parents may result in long-term implications for the health of offspring. Differences in diet appear to be a major driver of the maternal and subsequently milk and offspring microbiomes. Further prospective longitudinal cohort studies are needed to examine nutrition and the microbiome to better understand its implications. Additionally, trials involving dietary interventions in child-bearing age adults should be explored to improve the mother and child's risks for metabolic diseases.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David Taylor Hendrixson
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
26
|
Sones J, Balogh O. Body Condition and Fertility in Dogs. Vet Clin North Am Small Anim Pract 2023:S0195-5616(23)00067-0. [PMID: 37211441 DOI: 10.1016/j.cvsm.2023.04.005] [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: 05/23/2023]
Abstract
Ideal body condition and nutritional status in dogs is essential for quality of life, including reproductive health. Herein, we review the implications body condition, particularly fat, has on puberty, fertility, pregnancy, and parturition in dogs. Ideal body condition at puberty is necessary for dogs to achieve sexual maturity and reproduce. Moreover, over and under conditioned female dogs have increased risk of adverse pregnancy, parturition, and neonatal outcomes. Less is known about body condition and male dog fertility but some evidence is provided in this article. Finally, recommendations for maintaining an ideal body condition in intact adult dogs for optimal fertility are provided.
Collapse
Affiliation(s)
- Jennifer Sones
- Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA 70803, USA.
| | - Orsolya Balogh
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, 215 Duck Pond Drive, Blacksburg, VA 24061, USA.
| |
Collapse
|
27
|
Biddulph C, Holmes M, Tran TD, Kuballa A, Davies PSW, Koorts P, Maher J. Associations between Maternal Nutrition and the Concentrations of Human Milk Oligosaccharides in a Cohort of Healthy Australian Lactating Women. Nutrients 2023; 15:2093. [PMID: 37432220 PMCID: PMC10180645 DOI: 10.3390/nu15092093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 07/12/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are complex glycans associated with positive infant health outcomes. The concentrations of HMOs in the milk of lactating women are associated with substantial intra- and inter-individual differences and may be influenced by maternal physiological and/or nutrition-related factors. The primary aim of this study was to explore potential influences of short-term maternal diet and current body composition on HMO profiles in mature human milk. Milk samples were collected at 3-4 months postpartum from 101 healthy Australian women using standardised procedures, and analysed for macronutrients (lactose, fat, and protein). In addition, HMO concentrations were analysed using liquid-chromatography mass-spectrometry (LC-MS). Maternal dietary data were collected using three validated 24-h dietary recalls, and the body composition of a subgroup of mothers was assessed by DEXA scans (n = 30). Most (79%) of the women were secretor-positive. Individual nutrients were not significantly correlated with HMO concentrations after correction for multiple comparisons (p > 0.05), except for dietary folate intake. DEXA scans revealed no associations between HMO profiles and maternal body composition during established lactation. The study findings suggest a lack of clear and consistent associations between maternal nutrition and HMO concentrations in mature human milk from healthy lactating women with adequate dietary intake. The prevailing influence of genetic variation in lactating mothers may overshadow any impact of maternal nutritional and/or physiological status on HMO composition in mature human milk.
Collapse
Affiliation(s)
- Caren Biddulph
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
| | - Mark Holmes
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
- School of Health, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
| | - Trong D. Tran
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
| | - Anna Kuballa
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
- School of Health, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
| | - Peter S. W. Davies
- Child Health Research Centre, University of Queensland (UQ), St. Lucia, QLS 4072, Australia
| | - Pieter Koorts
- Department of Neonatology, Royal Brisbane and Women’s Hospital, Herston, QLS 4029, Australia
| | - Judith Maher
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
- School of Health, University of the Sunshine Coast, Maroochydore DC, QLS 4558, Australia
| |
Collapse
|
28
|
Neville MC, Demerath EW, Hahn-Holbrook J, Hovey RC, Martin-Carli J, McGuire MA, Newton ER, Rasmussen KM, Rudolph MC, Raiten DJ. Parental factors that impact the ecology of human mammary development, milk secretion, and milk composition-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 1. Am J Clin Nutr 2023; 117 Suppl 1:S11-S27. [PMID: 37173058 PMCID: PMC10232333 DOI: 10.1016/j.ajcnut.2022.11.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 05/15/2023] Open
Abstract
The goal of Working Group 1 in the Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN) Project was to outline factors influencing biological processes governing human milk secretion and to evaluate our current knowledge of these processes. Many factors regulate mammary gland development in utero, during puberty, in pregnancy, through secretory activation, and at weaning. These factors include breast anatomy, breast vasculature, diet, and the lactating parent's hormonal milieu including estrogen, progesterone, placental lactogen, cortisol, prolactin, and growth hormone. We examine the effects of time of day and postpartum interval on milk secretion, along with the role and mechanisms of lactating parent-infant interactions on milk secretion and bonding, with particular attention to the actions of oxytocin on the mammary gland and the pleasure systems in the brain. We then consider the potential effects of clinical conditions including infection, pre-eclampsia, preterm birth, cardiovascular health, inflammatory states, mastitis, and particularly, gestational diabetes and obesity. Although we know a great deal about the transporter systems by which zinc and calcium pass from the blood stream into milk, the interactions and cellular localization of transporters that carry substrates such as glucose, amino acids, copper, and the many other trace metals present in human milk across plasma and intracellular membranes require more research. We pose the question of how cultured mammary alveolar cells and animal models can help answer lingering questions about the mechanisms and regulation of human milk secretion. We raise questions about the role of the lactating parent and the infant microbiome and the immune system during breast development, secretion of immune molecules into milk, and protection of the breast from pathogens. Finally, we consider the effect of medications, recreational and illicit drugs, pesticides, and endocrine-disrupting chemicals on milk secretion and composition, emphasizing that this area needs much more research attention.
Collapse
Affiliation(s)
- Margaret C Neville
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA.
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, United States
| | - Jennifer Hahn-Holbrook
- Department of Psychological Sciences, University of California Merced, Merced, CA, United States
| | - Russell C Hovey
- Department of Animal Science, University of California Davis, Davis, CA, United States
| | - Jayne Martin-Carli
- Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Mark A McGuire
- Idaho Agricultural Experiment Station, University of Idaho, Moscow, ID, United States
| | - Edward R Newton
- Department of Obstetrics and Gynecology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Kathleen M Rasmussen
- Nancy Schlegel Meinig Professor of Maternal and Child Nutrition, Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Michael C Rudolph
- The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
29
|
Fan Y, Vinjamuri A, Tu D, Lebrilla CB, Donovan SM. Determinants of human milk oligosaccharides profiles of participants in the STRONG kids 2 cohort. Front Nutr 2023; 10:1105668. [PMID: 37057069 PMCID: PMC10086122 DOI: 10.3389/fnut.2023.1105668] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
IntroductionHuman milk oligosaccharides (HMOS) are indigestible carbohydrates that support infant development by establishing a healthy microbiota, preventing infectious diseases, and promoting immune and cognitive development. Individual HMOS have distinct functions based on their chemical structures. HMO profiles can vary largely among mothers, but the research on factors other than genetic background affecting HMO composition are limited.MethodsIn the present analysis, we examined the relationships between maternal characteristics and the HMO profiles of breastfeeding mothers (n = 392) in the STRONG kids 2 with the following demographic characteristics: average age: 30.8 y, 74.5% White, and 75.5% exclusively breastfeeding. Human milk samples were collected at 6 weeks postpartum and maternal information was obtained from self-reported surveys. Information on dietary intake changes since the participants have been breastfeeding was collected. HMO profiles were analyzed by high performance liquid chromatography coupled with mass spectrometry and secretor status was determined by the presence of four secretor markers [2′-fucosyllactose (2′-FL), LNFP I, LDFT, and TFLNH]. Spearmen correlation test was utilized to determine the relationships between individual HMOS and associations with maternal factors. Between-group differences in HMO relative abundances were examined with Kruskal-Wallis test.ResultsAmong all participants, 71.9% were secretors and 28.1% were non-secretors. The relative abundances of all HMOS differed (p < 0.05) by secretor status, with the exception for 6′-SL and 3′-SL. Positive correlations were observed among HMOS with similar structures, such as the 1,2-fucosylated HMOS. The abundances of selected HMOS were associated with maternal body weight, pregnancy complications, and dietary characteristics. Based on pre-pregnancy BMI, in all mothers, relative abundance of 3′-SL was significantly higher in overweight mothers than obese mothers (p = 0.013). In milk produced by non-secretor mothers, LNPF I + III abundances were greater in overweight than normal weight mothers (p = 0.020). Several HMO abundances were found to be associated with Gestational diabetes mellitus (GDM). Variations of HMO abundances were also observed with dietary food intake. In all mothers, egg consumption was positively correlated with LNT + LNnT (R = 0.13; p = 0.012) and cheese intake was positively associated with 2′-FL (R = 0.10; p = 0.046) and S-LNnH II (R = 0.11; p = 0.026) abundances.DiscussionHMO profiles were found to be associated with maternal characteristics and intake. Future research will investigate associations between HMOS and maternal and infant outcomes.
Collapse
Affiliation(s)
- Yuting Fan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, United States
| | - Anita Vinjamuri
- Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Diane Tu
- Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Carlito B. Lebrilla
- Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, United States
- *Correspondence: Sharon M. Donovan,
| |
Collapse
|
30
|
Nieto-Ruiz A, Cerdó T, Jordano B, Torres-Espínola FJ, Escudero-Marín M, García-Ricobaraza M, Bermúdez MG, García-Santos JA, Suárez A, Campoy C. Maternal weight, gut microbiota, and the association with early childhood behavior: the PREOBE follow-up study. Child Adolesc Psychiatry Ment Health 2023; 17:41. [PMID: 36945049 PMCID: PMC10031971 DOI: 10.1186/s13034-023-00589-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND AND AIM Maternal overweight and breastfeeding seem to have a significant impact on the gut microbiota colonization process, which co-occurs simultaneously with brain development and the establishment of the "microbiota-gut-brain axis", which potentially may affect behavior later in life. This study aimed to examine the influence of maternal overweight, obesity and/or gestational diabetes on the offspring behavior at 3.5 years of age and its association with the gut microbiota already established at 18 months of life. METHODS 156 children born to overweight (OV, n = 45), obese (OB, n = 40) and normoweight (NW, n = 71) pregnant women participating in the PREOBE study were included in the current analysis. Stool samples were collected at 18 months of life and gut microbiome was obtained by 16S rRNA gene sequencing. Behavioral problems were evaluated at 3.5 years by using the Child Behavior Checklist (CBCL). ANOVA, Chi-Square Test, ANCOVA, Spearman's correlation, logistic regression model and generalized linear model (GLM) were performed. RESULTS At 3.5 years of age, Children born to OV/OB mothers showed higher scores in behavioral problems than those born to NW mothers. Additionally, offspring born to OB mothers who developed gestational diabetes mellitus (GDM) presented higher scores in attention/deficit hyperactivity and externalizing problems than those born to GDM OV/NW mothers. Fusicatenibacter abundance found at 18 months of age was associated to lower scores in total, internalizing and pervasive developmental problems, while an unidentified genus within Clostridiales and Flavonifractor families abundance showed a positive correlation with anxiety/depression and somatic complaints, respectively. On the other hand, children born to mothers with higher BMI who were breastfed presented elevated anxiety, internalizing problems, externalizing problems and total problems scores; likewise, their gut microbiota composition at 18 months of age showed positive correlation with behavioral problems at 3.5 years: Actinobacteria abundance and somatic complaints and between Fusobacteria abundance and withdrawn behavior and pervasive developmental problems. CONCLUSIONS Our findings suggests that OV/OB and/or GDM during pregnancy is associated with higher behavioral problems scores in children at 3.5 years old. Additionally, associations between early life gut microbiota composition and later mental health in children was also found.
Collapse
Affiliation(s)
- Ana Nieto-Ruiz
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain
| | - Tomás Cerdó
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Belén Jordano
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain
- Clinical University Hospital San Cecilio. Paediatric Service, Granada, Spain
| | - Francisco J Torres-Espínola
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
| | - Mireia Escudero-Marín
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Neurosciences Institute Dr. Federico Oloriz - University of Granada. Health Sciences Technological Park, Avda. del Conocimiento, S/N., 18016, Granada, Spain
| | - María García-Ricobaraza
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain
| | - Mercedes G Bermúdez
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain
| | - José A García-Santos
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain
| | - Antonio Suárez
- Department of Biochemistry and Molecular Biology 2, School of Pharmacy, University of Granada, Granada, Spain
- Institute of Nutrition and Food Technology (INYTA), Biomedical Research Centre, University of Granada, Health Sciences Technological Park, Avda. del Conocimiento, S/N., 18016, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, Faculty of Medicine, University of Granada, Avda. Investigación 11, 18016, Granada, Spain.
- Biomedical Research Centre, EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18016, Granada, Spain.
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), San Cecilio University Hospital. Health Sciences Technological Park, 18016, Granada, Spain.
- Neurosciences Institute Dr. Federico Oloriz - University of Granada. Health Sciences Technological Park, Avda. del Conocimiento, S/N., 18016, Granada, Spain.
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's Node, Institute of Health Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
31
|
Baumgartel K, Stevens M, Vijayakumar N, Saint Fleur A, Prescott S, Groer M. The Human Milk Metabolome: A Scoping Literature Review. J Hum Lact 2023; 39:255-277. [PMID: 36924445 DOI: 10.1177/08903344231156449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
BACKGROUND Human milk is a complex source of nutrition and other bioactives that protects infants from disease, holding a lifetime of beneficial effects. The field of metabolomics provides a robust platform through which we can better understand human milk at a level rarely examined. RESEARCH AIM To Identify, describe, synthesize, and critically analyze the literature within the past 5 years related to the human milk metabolome. METHODS We conducted a scoping literature review and quality analysis of the recent science reflecting untargeted metabolomic approaches to examining human milk. We searched six databases using the terms "breast milk," "metabolome," "metabolite," and "human milk," Out of more than 1,069 abstracts, we screened and identified 22 articles that met our inclusion criteria. RESULTS We extracted data related to the study author, geographic location, research design, analyses, platform used, and results. We also extracted data related to human milk research activities, including collection protocol, infant/maternal considerations, and time. Selected studies focused on a variety of phenotypes, including maternal and infant disease. Investigators used varying approaches to evaluate the metabolome, and differing milk collection protocols were observed. CONCLUSION The human milk metabolome is informed by many factors-which may contribute to infant health outcomes-that have resulted in disparate milk metabolomic profiles. Standardized milk collection and storage procedures should be implemented to minimize degradation. Investigators may use our findings to develop research questions that test a targeted metabolomic approach.
Collapse
Affiliation(s)
| | - Monica Stevens
- College of Medicine, University of South Florida, Tampa, FL, USA
| | - Nisha Vijayakumar
- School of Public Health, University of South Florida, Tampa, FL, USA
| | | | | | - Maureen Groer
- College of Nursing, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
32
|
Song Y, Guo HL, Zhang MZ, Zhang ZL, Jin K, He QQ, Li H. lncRNA‑miRNA‑mRNA network in female offspring born from obese dams. Exp Ther Med 2023; 25:140. [PMID: 36845957 PMCID: PMC9947576 DOI: 10.3892/etm.2023.11839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 10/28/2022] [Indexed: 02/15/2023] Open
Abstract
Maternal obesity is associated with disturbance of lipid metabolism and obesity in offspring; however, the pathogenesis is still unclear. The present study elucidated the role of potential lipid metabolism-associated long non-coding RNA (lncRNA) and identified the pathways involved in mice born to obese dams. In the present study, maternal obesity was induced by feeding a high-fat diet for 10 weeks in female C57/BL6 mice, whereas control mice were fed a standard diet. All female mice mated with healthy male mice and were allowed to deliver spontaneously. The results demonstrated that female offspring from obese dams presented a tendency to become overweight in the first 8 weeks after birth; however, maternal obesity did not significantly alter the body weight of male offspring. RNA-sequencing analysis was performed on female offspring liver at 3 weeks old. Significantly dysregulated lncRNAs and downstream targets in female offspring liver were identified using bioinformatics analysis. lncRNA, microRNA (miRNA or miR) and mRNA expression levels in liver and AML12 cells were assessed using reverse transcription-quantitative PCR. A total of 8 upregulated and 17 downregulated lncRNAs were demonstrated in offspring from obese dams and lncRNA Lockd was indicated to be a key dysregulated lncRNA. Competing endogenous RNA (ceRNA) models suggested that the lncRNA Lockd/miR-582-5p/Elovl5 pathway was key for lipid metabolism in the liver of offspring from obese dams. Finally, small interfering RNA and miRNA inhibitor transfection was used to evaluate the ceRNA models in AML12 cells. Taken together, the results of the present study indicated that lncRNA Lockd-miR-582-5p-Elovl5 network may be disrupted in lipid metabolism and lead to obesity in the offspring of obese dams. This research will provide new insights into the molecular mechanism of obesity and lipid metabolism disorder.
Collapse
Affiliation(s)
- Yong Song
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, P.R. China,Institute of Preventive Medicine Information, Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei 430079, P.R. China
| | - Hong-Lin Guo
- School of Public Administration, South Central University for Nationalities, Wuhan, Hubei 430074, P.R. China
| | - Min-Zhe Zhang
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ze-Lin Zhang
- Department of Clinical Nutrition, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Ke Jin
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, Hubei 430023, P.R. China
| | - Qi-Qiang He
- Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong 518000, P.R. China
| | - Hui Li
- Medical Department, Taixing People's Hospital, Taizhou, Jiangsu 225400, P.R. China,Correspondence to: Miss Hui Li, Medical Department, Taixing People's Hospital, 1 Changzheng Road, Taixing, Taizhou, Jiangsu 225400, P.R. China
| |
Collapse
|
33
|
Rideout TC, Andreani GA, Pembroke J, Choudhary D, Browne RW, Mahmood S, Patel MS. Maternal Pea Protein Intake Provides Sex-Specific Protection against Dyslipidemia in Offspring from Obese Pregnancies. Nutrients 2023; 15:nu15040867. [PMID: 36839225 PMCID: PMC9968008 DOI: 10.3390/nu15040867] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Increased consumption of dietary pulse protein has been shown to assist in body weight regulation and improve a range of metabolic health outcomes. We investigated if the exchange of casein for yellow pea protein (YPPN) in an obese-inducing maternal diet throughout pregnancy and lactation offered protection against obesity and dyslipidemia in offspring. Sixty female Sprague Dawley rats were fed a low-calorie control diet (CON), a high-caloric obesity-inducing diet (with casein protein (CP), HC-CP), or an isocaloric/macronutrient-matched HC diet supplemented with YPPN isolate (HC-PPN) in pre-pregnancy, gestation, and lactation. Body weight (BW) and metabolic outcomes were assessed in male and female offspring at weaning and in adulthood after consuming the CON diet in the postnatal period. Consumption of the HC-PPN diet did not protect against maternal obesity but did improve reproductive success compared with the HC-CP group (72.7% versus 43.7%) and reduced total energy, fat, and protein in maternal milk. Male, but not female, offspring from mothers fed the HC-CP diet demonstrated hyperphagia, obesity, dyslipidemia, and hepatic triglyceride (TG) accumulation as adults compared with CON offspring. Isocaloric exchange of CP for YPPN in a high-calorie obese-inducing diet did not protect against obesity but did improve several aspects of lipid metabolism in adult male offspring including serum total cholesterol, LDL/VLDL cholesterol, triglycerides (TGs), and hepatic TG concentration. Our results suggest that the exchange of CP for YPPN in a maternal obese-inducing diet selectively protects male offspring from the malprogramming of lipid metabolism in adulthood.
Collapse
Affiliation(s)
- Todd C. Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
- Correspondence:
| | - Gabriella A. Andreani
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Jillian Pembroke
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Divya Choudhary
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Richard W. Browne
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Saleh Mahmood
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Mulchand S. Patel
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
34
|
Pintus R, Dessì A, Mussap M, Fanos V. Metabolomics can provide new insights into perinatal nutrition. Acta Paediatr 2023; 112:233-241. [PMID: 34487568 PMCID: PMC10078676 DOI: 10.1111/apa.16096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/13/2023]
Abstract
Perinatal nutrition is a key factor related to the Developmental Origin of Health and Disease hypothesis, which states that each and every event that happens during the periconceptional period and pregnancy can affect the health status of an individual. Metabolomics can be a very useful tool for gathering information about the effect of perinatal nutrition on both mothers and newborn infants. This non-systematic review focuses on the main metabolites detected by this technique, with regard to gestational diabetes, intrauterine growth restriction and breast milk. Conclusion. Nutrition, metabolome and microbiome interactions are gaining interest in the scientific community.
Collapse
Affiliation(s)
- Roberta Pintus
- Neonatal Intensive Care Unit, AOU Cagliari Department of Surgery, University of Cagliari, Cagliari, Italy
| | - Angelica Dessì
- Neonatal Intensive Care Unit, AOU Cagliari Department of Surgery, University of Cagliari, Cagliari, Italy
| | - Michele Mussap
- Neonatal Intensive Care Unit, AOU Cagliari Department of Surgery, University of Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, AOU Cagliari Department of Surgery, University of Cagliari, Cagliari, Italy
| |
Collapse
|
35
|
Gil-Kulik P, Leśniewski M, Bieńko K, Wójcik M, Więckowska M, Przywara D, Petniak A, Kondracka A, Świstowska M, Szymanowski R, Wilińska A, Wiliński M, Płachno BJ, Kostuch M, Rahnama-Hezavach M, Szuta M, Kwaśniewska A, Bogucka-Kocka A, Kocki J. Influence of Perinatal Factors on Gene Expression of IAPs Family and Main Factors of Pluripotency: OCT4 and SOX2 in Human Breast Milk Stem Cells-A Preliminary Report. Int J Mol Sci 2023; 24:ijms24032476. [PMID: 36768802 PMCID: PMC9917041 DOI: 10.3390/ijms24032476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Due to their therapeutic potential, mesenchymal stem cells are the subject of intensive research on the use of their potential in the treatment of, among others, neurodegenerative diseases or immunological diseases. They are among the newest in the field of medicine. The presented study aimed to evaluate the expression of eight genes from the IAP family and the gene regulating IAP-XAF1-in stem cells derived from human milk, using the qPCR method. The relationships between the expression of genes under study and clinical data, such as maternal age, maternal BMI, week of pregnancy in which the delivery took place, bodyweight of the newborn, the number of pregnancies and deliveries, and the time elapsed since delivery, were also analyzed. The research was carried out on samples of human milk collected from 42 patients hospitalized in The Clinic of Obstetrics and Perinatology of the Independent Public Clinical Hospital No. 4, in Lublin. The conducted research confirmed the expression of the following genes in the tested material: NAIP, BIRC2, BIRC3, BIRC5, BIRC6, BIRC8, XIAP, XAF1, OCT4 and SOX2. Moreover, several dependencies of the expression of individual genes on the maternal BMI (BIRC5, XAF1 and NAIP), the time since childbirth (BIRC5, BIRC6, XAF1 and NAIP), the number of pregnancies and deliveries (BIRC2, BIRC5, BIRC6 and XAF1), the manner of delivery (XAF1 and OCT4), preterm labor (BIRC6 and NAIP) were demonstrated. Additionally, we found positive relationships between gene expression of BIRC7, BIRC8 and XAF1 and the main factors of pluripotency: SOX2 and OCT4. This work is the first to investigate the expression of genes from the IAPs family in mother's milk stem cells.
Collapse
Affiliation(s)
- Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Michał Leśniewski
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Karolina Bieńko
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Monika Wójcik
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Marta Więckowska
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Dominika Przywara
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Alicja Petniak
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 11 Staszica Str., 20-081 Lublin, Poland
| | - Małgorzata Świstowska
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Rafał Szymanowski
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Agnieszka Wilińska
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Mateusz Wiliński
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Bartosz J. Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Faculty of Biology, Jagiellonian University in Kraków, 9 Gronostajowa St., 30-387 Cracow, Poland
| | - Marzena Kostuch
- Department of Neonatology, Independent Public Clinical Hospital No. 4, 8 Jaczewskiego St., 20-954 Lublin, Poland
| | - Mansur Rahnama-Hezavach
- Chair and Department of Dental Surgery, Medical University of Lublin, 6 Chodzki St., 20-093 Lublin, Poland
| | - Mariusz Szuta
- Chair of Oral Surgery, Jagiellonian University Medical College, 4 Montelupich St., 31-155 Kraków, Poland
| | - Anna Kwaśniewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 11 Staszica Str., 20-081 Lublin, Poland
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20–093 Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
- Correspondence:
| |
Collapse
|
36
|
Johnson KE, Heisel T, Allert M, Fürst A, Yerabandi N, Knights D, Jacobs KM, Lock EF, Bode L, Fields DA, Rudolph MC, Gale CA, Albert FW, Demerath EW, Blekhman R. Human milk variation is shaped by maternal genetics and impacts the infant gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525211. [PMID: 36747843 PMCID: PMC9900818 DOI: 10.1101/2023.01.24.525211] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Human milk is a complex mix of nutritional and bioactive components that provide complete nutrition for the infant. However, we lack a systematic knowledge of the factors shaping milk composition and how milk variation influences infant health. Here, we used multi-omic profiling to characterize interactions between maternal genetics, milk gene expression, milk composition, and the infant fecal microbiome in 242 exclusively breastfeeding mother-infant pairs. We identified 487 genetic loci associated with milk gene expression unique to the lactating mammary gland, including loci that impacted breast cancer risk and human milk oligosaccharide concentration. Integrative analyses uncovered connections between milk gene expression and infant gut microbiome, including an association between the expression of inflammation-related genes with IL-6 concentration in milk and the abundance of Bifidobacteria in the infant gut. Our results show how an improved understanding of the genetics and genomics of human milk connects lactation biology with maternal and infant health.
Collapse
Affiliation(s)
- Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Timothy Heisel
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mattea Allert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Annalee Fürst
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Nikhila Yerabandi
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Dan Knights
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Katherine M Jacobs
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal-Fetal Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Human Milk Institute (HMI) and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA
| | - David A Fields
- Department of Pediatrics, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, Department of Physiology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Cheryl A Gale
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
37
|
Varshney R, Das S, Trahan GD, Farriester JW, Mullen GP, Kyere-Davies G, Presby DM, Houck JA, Webb PG, Dzieciatkowska M, Jones KL, Rodeheffer MS, Friedman JE, MacLean PS, Rudolph MC. Neonatal intake of Omega-3 fatty acids enhances lipid oxidation in adipocyte precursors. iScience 2023; 26:105750. [PMID: 36590177 PMCID: PMC9800552 DOI: 10.1016/j.isci.2022.105750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 09/26/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Establishing metabolic programming begins during fetal and postnatal development, and early-life lipid exposures play a critical role during neonatal adipogenesis. We define how neonatal consumption of a low omega-6 to -3 fatty acid ratio (n6/n3 FA ratio) establishes FA oxidation in adipocyte precursor cells (APCs) before they become adipocytes. In vivo, APCs isolated from mouse pups exposed to the low n6/n3 FA ratio had superior FA oxidation capacity, elevated beige adipocyte mRNAs Ppargc1α, Ucp2, and Runx1, and increased nuclear receptor NR2F2 protein. In vitro, APC treatment with NR2F2 ligand-induced beige adipocyte mRNAs and increased mitochondrial potential but not mass. Single-cell RNA-sequencing analysis revealed low n6/n3 FA ratio yielded more mitochondrial-high APCs and linked APC NR2F2 levels with beige adipocyte signatures and FA oxidation. Establishing beige adipogenesis is of clinical relevance, because fat depots with energetically active, smaller, and more numerous adipocytes improve metabolism and delay metabolic dysfunction.
Collapse
Affiliation(s)
- Rohan Varshney
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - G. Devon Trahan
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob W. Farriester
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gregory P. Mullen
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gertrude Kyere-Davies
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - David M. Presby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Julie A. Houck
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Patricia G. Webb
- Department of Reproductive Science, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Kenneth L. Jones
- Department of Cell Biology and Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew S. Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Department of Comparative Medicine, Yale University, New Haven, CT, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paul S. MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Michael C. Rudolph
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
38
|
Lerin C, Collado MC, Isganaitis E, Arning E, Wasek B, Demerath EW, Fields DA, Bottiglieri T. Revisiting One-Carbon Metabolites in Human Breast Milk: Focus on S-Adenosylmethionine. Nutrients 2023; 15:282. [PMID: 36678154 PMCID: PMC9863976 DOI: 10.3390/nu15020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023] Open
Abstract
Breastfeeding is the gold standard for early nutrition. Metabolites from the one-carbon metabolism pool are crucial for infant development. The aim of this study is to compare the breast-milk one-carbon metabolic profile to other biofluids where these metabolites are present, including cord and adult blood plasma as well as cerebrospinal fluid. Breast milk (n = 142), cord blood plasma (n = 23), maternal plasma (n = 28), aging adult plasma (n = 91), cerebrospinal fluid (n = 92), and infant milk formula (n = 11) samples were analyzed by LC-MS/MS to quantify choline, betaine, methionine, S-adenosylmethionine, S-adenosylhomocysteine, total homocysteine, and cystathionine. Differences between groups were visualized by principal component analysis and analyzed by Kruskal-Wallis test. Correlation analysis was performed between one-carbon metabolites in human breast milk. Principal component analysis based on these metabolites separated breast milk samples from other biofluids. The S-adenosylmethionine (SAM) concentration was significantly higher in breast milk compared to the other biofluids and was absent in infant milk formulas. Despite many significant correlations between metabolites in one-carbon metabolism, there were no significant correlations between SAM and methionine or total homocysteine. Together, our data indicate a high concentration of SAM in breast milk, which may suggest a strong demand for this metabolite during infant early growth while its absence in infant milk formulas may indicate the inadequacy of this vital metabolic nutrient.
Collapse
Affiliation(s)
- Carles Lerin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Elvira Isganaitis
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erland Arning
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX 75204, USA
| | - Brandi Wasek
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX 75204, USA
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, The University of Minnesota School of Public Health, Minneapolis, MN 55455, USA
| | - David A. Fields
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX 75204, USA
| |
Collapse
|
39
|
Johnson W, Pereira SMP, Costa S, Baker JL, Norris T. The associations of maternal and paternal obesity with latent patterns of offspring BMI development between 7 and 17 years of age: pooled analyses of cohorts born in 1958 and 2001 in the United Kingdom. Int J Obes (Lond) 2023; 47:39-50. [PMID: 36357563 PMCID: PMC9834052 DOI: 10.1038/s41366-022-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE We aimed to 1) describe how the UK obesity epidemic reflects a change over time in the proportion of the population demonstrating adverse latent patterns of BMI development and 2) investigate the potential roles of maternal and paternal BMI in this secular process. METHODS We used serial BMI data between 7 and 17 years of age from 13220 boys and 12711 girls. Half the sample was born in 1958 and half in 2001. Sex-specific growth mixture models were developed. The relationships of maternal and paternal BMI and weight status with class membership were estimated using the 3-step BCH approach, with covariate adjustment. RESULTS The selected models had five classes. For each sex, in addition to the two largest normal weight classes, there were "normal weight increasing to overweight" (17% of boys and 20% of girls), "overweight increasing to obesity" (8% and 6%), and "overweight decreasing to normal weight" (3% and 6%) classes. More than 1-in-10 children from the 2001 birth cohort were in the "overweight increasing to obesity" class, compared to less than 1-in-30 from the 1958 birth cohort. Approximately 75% of the mothers and fathers of this class had overweight or obesity. When considered together, both maternal and paternal BMI were associated with latent class membership, with evidence of negative departure from additivity (i.e., the combined effect of maternal and paternal BMI was smaller than the sum of the individual effects). The odds of a girl belonging to the "overweight increasing to obesity" class (compared to the largest normal weight class) was 13.11 (8.74, 19.66) times higher if both parents had overweight or obesity (compared to both parents having normal weight); the equivalent estimate for boys was 9.01 (6.37, 12.75). CONCLUSIONS The increase in obesity rates in the UK over more than 40 years has been partly driven by the growth of a sub-population demonstrating excess BMI gain during adolescence. Our results implicate both maternal and paternal BMI as correlates of this secular process.
Collapse
Affiliation(s)
- William Johnson
- grid.6571.50000 0004 1936 8542School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Snehal M. Pinto Pereira
- grid.83440.3b0000000121901201UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - Silvia Costa
- grid.6571.50000 0004 1936 8542School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jennifer L. Baker
- grid.411702.10000 0000 9350 8874Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Tom Norris
- grid.83440.3b0000000121901201UCL Division of Surgery & Interventional Science, University College London, London, UK
| |
Collapse
|
40
|
Ross MG, Kobayashi K, Han G, Desai M. Modulation of Milk and Lipid Synthesis and Secretion in a3-Dimensional Mouse Mammary Epithelial Cell Culture Model: Effects of Palmitate and Orlistat. Nutrients 2022; 14:4948. [PMID: 36500977 PMCID: PMC9739267 DOI: 10.3390/nu14234948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Human milk synthesis is impacted by maternal diet, serum composition, and substrate uptake and synthesis by mammary epithelial cells (MECs). The milk of obese/high-fat-diet women has an increased fat content, which promote excess infant weight gain and the risk of childhood/adult obesity. Yet, the knowledge of milk synthesis regulation is limited, and there are no established approaches to modulate human milk composition. We established a 3-dimensional mouse MEC primary culture that recreates the milk production pathway and tested the effects of the major saturated fatty acid in human milk (palmitate) and a lipoprotein lipase inhibitor (orlistat) on triglyceride production. Positive immunostaining confirmed the presence of milk protein and intracellular lipid including milk globules in the cytoplasm and extracellular space. The treatment with palmitate activated "milk" production by MECs (β-casein) and the lipid pathway (as evident by increased protein and mRNA expression). Consistent with these cellular changes, there was increased secretion of milk protein and triglyceride in MEC "milk". The treatment with orlistat suppressed milk triglyceride production. Palmitate increased milk and lipid synthesis, partly via lipoprotein lipase activation. These findings demonstrate the ability to examine MEC pathways of milk production via both protein and mRNA and to modulate select pathways regulating milk composition in MEC culture.
Collapse
Affiliation(s)
- Michael G. Ross
- The Lundquist Institute at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA 90502, USA
- Department of Obstetrics and Gynecology, Charles R. Drew University, Los Angeles, CA 90059, USA
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| | - Guang Han
- The Lundquist Institute at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Mina Desai
- The Lundquist Institute at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA 90502, USA
| |
Collapse
|
41
|
Overgaard Poulsen K, Astono J, Jakobsen RR, Uldbjerg N, Fuglsang J, Nielsen DS, Sundekilde UK. Influence of maternal body mass index on human milk composition and associations to infant metabolism and gut colonisation: MAINHEALTH - a study protocol for an observational birth cohort. BMJ Open 2022; 12:e059552. [PMID: 36323479 PMCID: PMC9639067 DOI: 10.1136/bmjopen-2021-059552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Human milk provides all macronutrients for growth, bioactive compounds, micro-organisms and immunological components, which potentially interacts with and primes infant growth and, development, immune responses and the gut microbiota of the new-born. Infants with an overweight mother are more likely to become overweight later in life and overweight has been related to the gut microbiome. Therefore, it is important to investigate the mother-milk-infant triad as a biological system and if the maternal weight status influences the human milk composition, infant metabolism and gut microbiome. METHODS AND ANALYSIS This study aims to include 200 mother-infant dyads stratified into one of three body mass index (BMI) categories based on mother's prepregnancy BMI. Multiomics analyses include metabolomics, proteomics, glycomics and microbiomics methods, aiming to characterise human milk from the mothers and further relate the composition to infant gut microbiota and its metabolic impact in the infant. Infant gut microbiota is analysed using 16S sequencing of faeces samples. Nuclear magnetic resonance and mass spectrometry are used for the remaining omics analysis. We investigate whether maternal pre-pregnancy BMI results in a distinct human milk composition that potentially affects the initial priming of the infant's gut environment and metabolism early in life. ETHICS AND DISSEMINATION The Central Denmark Region Committees on Health Research Ethics has approved the protocol (J-nr. 1-10-72-296-18). All participants have before inclusion signed informed consent and deputy informed consent in accordance with the Declaration of Helsinki II. Results will be disseminated to health professionals including paediatricians, research community, nutritional policymakers, industry and finally the public. The scientific community will be informed via peer-reviewed publications and presentations at scientific conferences, the industry will be invited for meetings, and the public will be informed via reports in science magazines and the general press. Data cleared for personal data, will be deposited at public data repositories. TRIAL REGISTRATION NUMBER Danish regional committee of the Central Jutland Region, journal number: 1-10-72-296-18, version 6.Danish Data Protection Agency, journal number: 2016-051-000001, 1304. CLINICALTRIALS gov, identifier: NCT05111990.
Collapse
Affiliation(s)
- Katrine Overgaard Poulsen
- Department of Food Science, Aarhus University, Aarhus N, Denmark
- Sino-Danish Centre for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Julie Astono
- Department of Food Science, Aarhus University, Aarhus N, Denmark
| | | | - Niels Uldbjerg
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Jens Fuglsang
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | | | | |
Collapse
|
42
|
Michońska I, Łuszczki E, Zielińska M, Oleksy Ł, Stolarczyk A, Dereń K. Nutritional Programming: History, Hypotheses, and the Role of Prenatal Factors in the Prevention of Metabolic Diseases-A Narrative Review. Nutrients 2022; 14:4422. [PMID: 36297106 PMCID: PMC9607048 DOI: 10.3390/nu14204422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Childhood obesity and the numerous lifestyle diseases associated with it are undoubtedly among the key problems in modern medicine and public health. However, this problem concerns not only the present or immediate future, but also the longer term. Adult health is fundamentally shaped in the first years of life and in the fetal period. The preconceptual period, which is responsible for the proper preparation of the internal environment for the life and development of the fetus during pregnancy, is also significant. A special role in describing the phenomenon of conditioning the metabolism of the new human being is now attributed to the theory of nutritional programming. Research in this area was pioneered by David Barker, who put forward the theory of the "stunted phenotype" and described the relationship between a child's birth weight, which is largely a consequence of the mother's feeding behaviour, and diseases such as ischaemic heart disease, type 2 diabetes (T2D), dyslipidemia, or high blood pressure. This narrative review aims to provide an overview of the history, theory, and prenatal mechanisms involved in nutritional programming and its relationship to childhood obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Izabela Michońska
- Institute of Health Sciences, College of Medical Sciences, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Edyta Łuszczki
- Institute of Health Sciences, College of Medical Sciences, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Magdalena Zielińska
- Institute of Health Sciences, College of Medical Sciences, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Łukasz Oleksy
- Faculty of Health Sciences, Department of Physiotherapy, Jagiellonian University Medical College Krakow, 31-008 Krakow, Poland
| | - Artur Stolarczyk
- Orthopedic and Rehabilitation Department, Medical Faculty, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Katarzyna Dereń
- Institute of Health Sciences, College of Medical Sciences, University of Rzeszow, 35-959 Rzeszow, Poland
| |
Collapse
|
43
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [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: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
44
|
Poulsen KO, Meng F, Lanfranchi E, Young JF, Stanton C, Ryan CA, Kelly AL, Sundekilde UK. Dynamic Changes in the Human Milk Metabolome Over 25 Weeks of Lactation. Front Nutr 2022; 9:917659. [PMID: 35911093 PMCID: PMC9331903 DOI: 10.3389/fnut.2022.917659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022] Open
Abstract
Human milk (HM) provides essential nutrition for ensuring optimal infant growth and development postpartum. Metabolomics offers insight into the dynamic composition of HM. Studies have reported the impact of lactation stage, maternal genotype, and gestational age on HM metabolome. However, the majority of the studies have considered changes within the first month of lactation or sampled with large intervals. This leaves a gap in the knowledge of progressing variation in HM composition beyond the first month of lactation. The objective of this study was to investigate whether the HM metabolome from mothers with term deliveries varies beyond 1 month of lactation, during the period in which HM is considered fully mature. Human milk samples (n = 101) from 59 mothers were collected at weeks 1-2, 3-5, 7-9, and 20-25 postpartum and analyzed using 1H nuclear magnetic resonance spectroscopy. Several metabolites varied over lactation and exhibited dynamic changes between multiple time points. Higher levels of HM oligosaccharides, cis-aconitate, O-phosphocholine, O-acetylcarnitine, gluconate, and citric acid were observed in early lactation, whereas later in lactation, levels of lactose, 3-fucosyllactose, glutamine, glutamate, and short- and medium-chain fatty acids were increased. Notably, we demonstrate that the HM metabolome is dynamic during the period of maturity.
Collapse
Affiliation(s)
- Katrine Overgaard Poulsen
- Department of Food Science, Aarhus University, Aarhus, Denmark
- Sino-Danish Center for Education and Research, Aarhus, Denmark
| | - Fanyu Meng
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Elisa Lanfranchi
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- ACIB – Austrian Centre of Industrial Biotechnology, Graz, Austria
| | | | | | - C. Anthony Ryan
- Brookfield School of Medicine and Health, University College Cork, Cork, Ireland
| | - Alan L. Kelly
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | | |
Collapse
|
45
|
Maternal Predictors of Breast Milk Plasmalogens and Associations with Infant Body Composition and Neurodevelopment. Clin Ther 2022; 44:998-1009. [DOI: 10.1016/j.clinthera.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/06/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
|
46
|
Gregg B, Ellsworth L, Pavela G, Shah K, Berger PK, Isganaitis E, VanOmen S, Demerath EW, Fields DA. Bioactive compounds in mothers milk affecting offspring outcomes: A narrative review. Pediatr Obes 2022; 17:e12892. [PMID: 35060344 PMCID: PMC9177518 DOI: 10.1111/ijpo.12892] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/07/2021] [Accepted: 01/03/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Compared to the exhaustive study of transgenerational programming of obesity and diabetes through exposures in the prenatal period, postnatal programming mechanisms are understudied, including the potential role of breast milk composition linking maternal metabolic status (body mass index and diabetes) and offspring growth, metabolic health and future disease risk. METHODS This narrative review will principally focus on four emergent bioactive compounds [microRNA's (miRNA), lipokines/signalling lipids, small molecules/metabolites and fructose] that, until recently were not known to exist in breast milk. The objective of this narrative review is to integrate evidence across multiple fields of study that demonstrate the importance of these compositional elements of breast milk during lactation and the subsequent effect of breast milk components on the health of the infant. RESULTS Current knowledge on the presence of miRNA's, lipokines/signalling lipids, small molecules/metabolites and fructose in breast milk and their associations with infant outcomes is compelling, but far from resolved. Two themes emerge: (1) maternal metabolic phenotypes are associated with these bioactives and (2) though existing in milk at low concentrations, they are also associated with offspring growth and body composition. CONCLUSION Breast milk research is gaining momentum though we must remain focused on understanding how non-nutritive bioactive components are affected by the maternal phenotype, how they subsequently impact infant outcomes. Though early, there is evidence to suggest fructose is associated with fat mass in the 1st months of life whereas 12,13 diHOME (brown fat activator) and betaine are negatively associated with early adiposity and growth.
Collapse
Affiliation(s)
- Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI, USA
| | - Lindsay Ellsworth
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gregory Pavela
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kruti Shah
- Department of Pediatrics, Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paige K. Berger
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Elvira Isganaitis
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA (USA)
| | - Sheri VanOmen
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - David A. Fields
- Department of Pediatrics, Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Correspondence to: Address: University of Oklahoma Health Sciences Center, 1200 Children's Avenue Suite 4500, Oklahoma City, OK73104, USA
| |
Collapse
|
47
|
The association of pre-pregnancy BMI on leptin, ghrelin, adiponectin and insulin-like growth factor-1 in breast milk: a case-control study. Br J Nutr 2022; 127:1675-1681. [PMID: 34348809 PMCID: PMC9201832 DOI: 10.1017/s0007114521002932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The nutrient composition of breast milk alters during lactation, and maternal BMI adds more intricacy into its complexity. We aimed to compare leptin, ghrelin, adiponectin and insulin-like growth factor-1 (IGF-1) levels of pre-feed and post-feed breast milk in mothers with obesity and normal weight, and tried to determine their effects on infants' growth over weight for length z-score. Twenty obese and twenty normal weight mothers with 2-month-old infants were enrolled in this case-control study. Five millilitre pre-feed breast milk and 5 ml post-feed breast milk were collected. Breast milk leptin, ghrelin, adiponectin and IGF-1 were measured by commercial kits. The pre-feed breast milk of mothers with obesity had significantly higher levels of ghrelin than mothers with normal weight (P = 0·025), whereas the post-feed breast milk of mothers with normal weight had higher levels of adiponectin than the mothers with obesity (P = 0·010). No significant differences were observed in leptin and IGF-1 levels between the two groups. Post-feed breast milk IGF-1 levels of mothers with obesity were correlated with infant's weight for length z-score at 2 months (r -0·476; P = 0·034). In linear regression models, parity affected the ghrelin in pre-feed breast milk (P = 0·025). Our results revealed that maternal pre-pregnancy BMI was associated with breast milk components.
Collapse
|
48
|
Beyond the BMI: Validity and Practicality of Postpartum Body Composition Assessment Methods during Lactation: A Scoping Review. Nutrients 2022; 14:nu14112197. [PMID: 35683995 PMCID: PMC9182963 DOI: 10.3390/nu14112197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
The assessment of body composition during lactation is an important indicator of maternal nutritional status, which is central to the overall health of the mother and child. The lactating woman’s nutritional status potentially impacts on breastmilk composition and the process of lactation itself. The purpose of this scoping review was to synthesize comparative studies that sought to validate various body composition assessment techniques for use in lactating women in the postpartum period. Using the PRISMA-ScR guidelines, a comprehensive, systematic literature search was conducted using Scopus, Web of Science, and PubMed. Eight comparative studies were included in the review, with data from 320 postpartum women. The design methodologies varied substantially across studies, and included a range of simple techniques to advanced multi-compartment models for assessing body composition. The validity and reliability of measurement tools must be considered alongside issues of safety, practicality, and appropriateness to guide the research design when applied to lactating women.
Collapse
|
49
|
Jantscher-Krenn E, von Schirnding L, Trötzmüller M, Köfeler H, Kurtovic U, Fluhr H, Müller A, Bagci S. Human Milk Oligosaccharides Are Present in Amniotic Fluid and Show Specific Patterns Dependent on Gestational Age. Nutrients 2022; 14:nu14102065. [PMID: 35631205 PMCID: PMC9146373 DOI: 10.3390/nu14102065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Human milk oligosaccharides (HMOs) are already found in maternal circulation in early pregnancy, changing with gestational age. HMOs are also present in cord blood and amniotic fluid (AF). We aimed to assess HMO profiles in AF over the course of gestation. (2) Methods: AF was collected during diagnostic amniocentesis, fetal surgery, or C-section from 77 women with a gestational age of ranging from 14.3 to 40.9 weeks. Samples were analysed using high performance liquid chromatography with fluorescence detection. (3) Results: We found lactose and up to 16 HMO structures in all AF samples investigated, starting at 14 weeks of gestation. Overall, 3′-sialyllactose (3′SL) and 2′-fucosyllactose (2′FL) were the most abundant HMOs. Individual and total HMO concentrations were significantly positively correlated with gestational age. HMO composition also changed between early, mid- and late pregnancy, with relative concentrations of 3′SL significantly decreasing (44%, 25%, 24%) and 2′FL increasing (7%, 13%, 21%), respectively. (4) Conclusion: Our study shows that HMOs are already present in AF early in pregnancy. This demonstrates extensive contact of the fetus with a broad variety of HMOs, suggesting roles for HMOs in fetal tissue development during the time course of pregnancy.
Collapse
Affiliation(s)
- Evelyn Jantscher-Krenn
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (U.K.); (H.F.)
- BioTechMed, 8010 Graz, Austria;
- Correspondence: (E.J.-K.); (S.B.); Tel.: +43-316-385-80076 (E.J.-K.); +49-228-287-37834 (S.B.)
| | - Lara von Schirnding
- Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, D-53113 Bonn, Germany; (L.v.S.); (A.M.)
| | - Martin Trötzmüller
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, 8036 Graz, Austria;
| | - Harald Köfeler
- BioTechMed, 8010 Graz, Austria;
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, 8036 Graz, Austria;
| | - Una Kurtovic
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (U.K.); (H.F.)
| | - Herbert Fluhr
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (U.K.); (H.F.)
| | - Andreas Müller
- Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, D-53113 Bonn, Germany; (L.v.S.); (A.M.)
| | - Soyhan Bagci
- Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, D-53113 Bonn, Germany; (L.v.S.); (A.M.)
- Correspondence: (E.J.-K.); (S.B.); Tel.: +43-316-385-80076 (E.J.-K.); +49-228-287-37834 (S.B.)
| |
Collapse
|
50
|
Weighted analysis of 2'-fucosylactose, 3-fucosyllactose, lacto-N-tetraose, 3'-sialyllactose, and 6'-sialyllactose concentrations in human milk. Food Chem Toxicol 2022; 163:112877. [PMID: 35304182 DOI: 10.1016/j.fct.2022.112877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 11/22/2022]
Abstract
Over 150 human milk oligosaccharides (HMOs) have been identified and their concentrations in human milk vary depending on Secretor and Lewis blood group status, environmental and geographical factors, lactation stage, gestational period, and maternal health. Quantitation of HMOs in human milk has been the focus of numerous studies, however, comprehensive and weighted statistical analyses of their levels in human milk are lacking. Therefore, weighted means, standard deviations, medians, interquartile ranges, and 90th percentiles for 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL), lacto-N-tetraose (LNT), 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL) were calculated using random sampling and the levels of these HMOs in human milk reported in the literature. Probability distributions of the reported levels were also constructed. Although the levels reported in the published studies varied, the weighted means for 2'-FL, 3-FL, LNT, 3'-SL, and 6'-SL were calculated to be 2.58, 0.57, 0.94, 0.28, and 0.39 g/L, respectively, which are consistent with those that have been previously determined in other systematic analyses. Likely due to the use of weighting, the 90th percentiles were greater than the 95% confidence limits that have been previously calculated. Our study therefore provides accurate and important statistical data to help support the level of appropriate HMO supplementation in infant formula.
Collapse
|