1
|
Albrecht M, Worthmann A, Heeren J, Diemert A, Arck PC. Maternal lipids in overweight and obesity: implications for pregnancy outcomes and offspring's body composition. Semin Immunopathol 2025; 47:10. [PMID: 39841244 PMCID: PMC11754334 DOI: 10.1007/s00281-024-01033-6] [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: 08/12/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Overweight and obesity (OWO) are linked to dyslipidemia and low-grade chronic inflammation, which is fueled by lipotoxicity and oxidative stress. In the context of pregnancy, maternal OWO has long been known to negatively impact on pregnancy outcomes and maternal health, as well as to imprint a higher risk for diseases in offspring later in life. Emerging research suggests that individual lipid metabolites, which collectively form the lipidome, may play a causal role in the pathogenesis of OWO-related diseases. This can be applied to the onset of pregnancy complications such as gestational diabetes mellitus (GDM) and hypertensive disorders of pregnancy (HDP), which in fact occur more frequently in women affected by OWO. In this review, we summarize current knowledge on maternal lipid metabolites in pregnancy and highlight associations between the maternal lipidome and the risk to develop GDM, HDP and childhood OWO. Emerging data underpin that dysregulations in maternal triglyceride, phospholipid and polyunsaturated fatty acid (PUFA) metabolism may play a role in modulating the risk for adverse pregnancy outcomes and childhood OWO, but it is yet premature to convert currently available insights into clinical guidelines. Well-designed large-scale lipidomic studies, combined with translational approaches including animal models of obesity, will likely facilitate the recognition of underling pathways of OWO-related pregnancy complications and child's health outcomes, based on which clinical guidelines and recommendations can be updated.
Collapse
Affiliation(s)
- Marie Albrecht
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany.
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Jörg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Clara Arck
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Gyllenhammer LE, Boyle KE. New Frontiers: Umbilical Cord Mesenchymal Stem Cells Uncover Developmental Roots and Biological Underpinnings of Obesity Susceptibility. Curr Obes Rep 2025; 14:10. [PMID: 39814984 PMCID: PMC11735562 DOI: 10.1007/s13679-024-00599-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE OF REVIEW To review evidence supporting human umbilical cord mesenchymal stem cells (UC-MSC) as an innovative model system advancing obesity precision medicine. RECENT FINDINGS Obesity prevalence is increasing rapidly and exposures during fetal development can impact individual susceptibility to obesity. UC-MSCs exhibit heterogeneous phenotypes associated with maternal exposures and predictive of child cardiometabolic outcomes. This recent evidence supports UC-MSCs as a precision model serving three purposes: (1) as a mechanistic tool to interrogate biological underpinnings of obesity in human studies, (2) as a sensitive index of early life causes and determinants of obesity, and (3) as a marker and transducer of susceptibility, highlighting populations most at risk for future obesity. Data from UC-MSCs emphasize nutrient sensing and lipid partitioning as phenotypes most relevant to neonatal and early childhood adiposity and implicate a role for these cell-autonomous features of mesodermal tissues in the biological underpinnings of obesity.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, USA.
| | - Kristen E Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA.
| |
Collapse
|
3
|
Martínez Velasco IG, Guillén González MA, Jiménez López R, Arciniega Mancilla O. [Omega-3 and preterm birth: Deciphering the pieces of the puzzle for its prevention]. NUTR HOSP 2024; 41:1317-1318. [PMID: 39512023 DOI: 10.20960/nh.05422] [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: 11/15/2024] Open
Abstract
Introduction
Collapse
Affiliation(s)
| | | | - Román Jiménez López
- Servicio de Obstetricia. Hospital Comunitario de Tecomatlán. Servicios de Salud del Estado de Puebla
| | | |
Collapse
|
4
|
Gsoellpointner M, Thanhaeuser M, Kornsteiner-Krenn M, Eibensteiner F, Ristl R, Jilma B, Brandstetter S, Berger A, Haiden N. Macronutrient Intake during Complementary Feeding in Very Low Birth Weight Infants Comparing Early and Late Introduction of Solid Foods: A Secondary Outcome Analysis. Nutrients 2024; 16:3422. [PMID: 39408387 PMCID: PMC11478545 DOI: 10.3390/nu16193422] [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: 08/29/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Very low birth weight (VLBW) infants may require enhanced nutrition, even during complementary feeding. However, there are limited data on macronutrient intake during this period, particularly concerning the individual timing of the introduction of solid foods in a representative VLBW infant population. Methods: This prospective observational study analyzed macronutrient intake in VLBW infants with a gestational age < 32 weeks based on whether solid foods were introduced early (<17 weeks corrected age (CA)) or late (≥17 weeks corrected age) Nutritional intake was analyzed using a 24 h recall at 6 weeks CA and 3-day dietary records at 12 weeks, 6, 9, and 12 months CA. Results: In total, 115 infants were assigned to the early and 82 to the late group. The timing of solid food introduction did not affect macronutrient intake, except for a lower fat and higher carbohydrate intake (% of energy) in the early group at 12 weeks and 6 months CA: early vs. late, fat-12 weeks: 47.0% vs. 49.0%, 6 months: 39.2% vs. 43.3%; carbohydrates-12 weeks: 44.9% vs. 43.2%, 6 months: 51.3% vs. 48.0%. Apart from docosahexaenoic acid (DHA) and arachidonic acid (AA), dietary intake recommendations were met in both groups. While nutrient intakes varied significantly between breastfed and formula-fed infants, those with comorbidities exhibited similar nutrient intake levels compared to those without. Conclusions: Our findings suggest adequate macronutrient intakes in VLBW infants irrespective of the timing of solid introduction. However, there is a notable need to enhance dietary intakes of DHA and AA. Future research is crucial to assess whether current nutrient intakes are sufficient for VLBW infants with comorbidities.
Collapse
Affiliation(s)
- Melanie Gsoellpointner
- Department of Neonatology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
| | - Margarita Thanhaeuser
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (M.T.); (M.K.-K.); (F.E.); (S.B.); (A.B.)
| | - Margit Kornsteiner-Krenn
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (M.T.); (M.K.-K.); (F.E.); (S.B.); (A.B.)
| | - Fabian Eibensteiner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (M.T.); (M.K.-K.); (F.E.); (S.B.); (A.B.)
| | - Robin Ristl
- Center for Medical Data Science, Medical University of Vienna, 1090 Vienna, Austria;
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Sophia Brandstetter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (M.T.); (M.K.-K.); (F.E.); (S.B.); (A.B.)
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (M.T.); (M.K.-K.); (F.E.); (S.B.); (A.B.)
| | - Nadja Haiden
- Department of Neonatology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
| |
Collapse
|
5
|
Das S, Varshney R, Farriester JW, Kyere-Davies G, Martinez AE, Hill K, Kinter M, Mullen GP, Nagareddy PR, Rudolph MC. NR2F2 Reactivation in Early-life Adipocyte Stem-like Cells Rescues Adipocyte Mitochondrial Oxidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.611047. [PMID: 39314382 PMCID: PMC11419096 DOI: 10.1101/2024.09.09.611047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
In humans, perinatal exposure to an elevated omega-6 (n6) relative to omega-3 (n3) Fatty Acid (FA) ratio is associated with the likelihood of childhood obesity. In mice, we show perinatal exposure to excessive n6-FA programs neonatal Adipocyte Stem-like cells (ASCs) to differentiate into adipocytes with lower mitochondrial nutrient oxidation and a propensity for nutrient storage. Omega-6 FA exposure reduced fatty acid oxidation (FAO) capacity, coinciding with impaired induction of beige adipocyte regulatory factors PPARγ, PGC1α, PRDM16, and UCP1. ASCs from n6-FA exposed pups formed adipocytes with increased lipogenic genes in vitro, consistent with an in vivo accelerated adipocyte hypertrophy, greater triacylglyceride accumulation, and increased % body fat. Conversely, n6-FA exposed pups had impaired whole animal 13C-palmitate oxidation. The metabolic nuclear receptor, NR2F2, was suppressed in ASCs by excess n6-FA intake preceding adipogenesis. ASC deletion of NR2F2, prior to adipogenesis, mimicked the reduced FAO capacity observed in ASCs from n6-FA exposed pups, suggesting that NR2F2 is required in ASCs for robust beige regulator expression and downstream nutrient oxidation in adipocytes. Transiently re-activating NR2F2 with ligand prior to differentiation in ASCs from n6-FA exposed pups, restored their FAO capacity as adipocytes by increasing the PPARγ-PGC1α axis, mitochondrial FA transporter CPT1A, ATP5 family synthases, and NDUF family Complex I proteins. Our findings suggest that excessive n6-FA exposure early in life dampens an NR2F2-mediated induction of beige adipocyte regulators, resulting in metabolic programming that is shifted towards nutrient storage.
Collapse
Affiliation(s)
- Snehasis Das
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Rohan Varshney
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Jacob W. Farriester
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Gertrude Kyere-Davies
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Alexandrea E. Martinez
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Kaitlyn Hill
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Gregory P. Mullen
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Prabhakara R. Nagareddy
- Deptartment of Internal Medicine, Cardiovascular Section, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Michael C. Rudolph
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Omachi DO, Aryee ANA, Onuh JO. Functional Lipids and Cardiovascular Disease Reduction: A Concise Review. Nutrients 2024; 16:2453. [PMID: 39125334 PMCID: PMC11314407 DOI: 10.3390/nu16152453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Functional lipids are dietary substances that may have an impact on human health by lowering the risk of chronic illnesses and enhancing the quality of life. Numerous functional lipids have been reported to have potential health benefits in the prevention, management, and treatment of cardiovascular disease, the leading cause of death in the United States. However, there is still insufficient and contradictory information in the literature about their effectiveness and associated mechanisms of action. The objective of this review, therefore, is to evaluate available literature regarding these functional lipids and their health benefits. Various studies have been conducted to understand the links between functional lipids and the prevention and treatment of chronic diseases. Recent studies on phytosterols have reported that CLA, medium-chain triglycerides, and omega-3 and 6 fatty acids have positive effects on human health. Also, eicosanoids, which are the metabolites of these fatty acids, are produced in relation to the ratio of omega-3 to omega-6 polyunsaturated fatty acids and may modulate disease conditions. These functional lipids are available either in dietary or supplement forms and have been proven to be efficient, accessible, and inexpensive to be included in the diet. However, further research is required to properly elucidate the dosages, dietary intake, effectiveness, and their mechanisms of action in addition to the development of valid disease biomarkers and long-term effects in humans.
Collapse
Affiliation(s)
- Deborah O. Omachi
- Department of Food and Nutritional Sciences, Tuskegee University, 1200 W. Montgomery Rd, Tuskegee, AL 36088, USA;
| | - Alberta N. A. Aryee
- Food Science and Biotechnology Program, Department of Human Ecology, Delaware State University, 1200 Dupont Highway, Dover, DE 19901, USA;
| | - John O. Onuh
- Department of Food and Nutritional Sciences, Tuskegee University, 1200 W. Montgomery Rd, Tuskegee, AL 36088, USA;
| |
Collapse
|
7
|
Martemucci G, Khalil M, Di Luca A, Abdallah H, D’Alessandro AG. Comprehensive Strategies for Metabolic Syndrome: How Nutrition, Dietary Polyphenols, Physical Activity, and Lifestyle Modifications Address Diabesity, Cardiovascular Diseases, and Neurodegenerative Conditions. Metabolites 2024; 14:327. [PMID: 38921462 PMCID: PMC11206163 DOI: 10.3390/metabo14060327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Several hallmarks of metabolic syndrome, such as dysregulation in the glucose and lipid metabolism, endothelial dysfunction, insulin resistance, low-to-medium systemic inflammation, and intestinal microbiota dysbiosis, represent a pathological bridge between metabolic syndrome and diabesity, cardiovascular, and neurodegenerative disorders. This review aims to highlight some therapeutic strategies against metabolic syndrome involving integrative approaches to improve lifestyle and daily diet. The beneficial effects of foods containing antioxidant polyphenols, intestinal microbiota control, and physical activity were also considered. We comprehensively examined a large body of published articles involving basic, animal, and human studie, as well as recent guidelines. As a result, dietary polyphenols from natural plant-based antioxidants and adherence to the Mediterranean diet, along with physical exercise, are promising complementary therapies to delay or prevent the onset of metabolic syndrome and counteract diabesity and cardiovascular diseases, as well as to protect against neurodegenerative disorders and cognitive decline. Modulation of the intestinal microbiota reduces the risks associated with MS, improves diabetes and cardiovascular diseases (CVD), and exerts neuroprotective action. Despite several studies, the estimation of dietary polyphenol intake is inconclusive and requires further evidence. Lifestyle interventions involving physical activity and reduced calorie intake can improve metabolic outcomes.
Collapse
Affiliation(s)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | - Alessio Di Luca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (A.D.L.); (A.G.D.)
| | - Hala Abdallah
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | | |
Collapse
|
8
|
Pacyga DC, Papandonatos GD, Rosas L, Whalen J, Smith S, Park JS, Gardiner JC, Braun JM, Schantz SL, Strakovsky RS. Associations of per- and polyfluoroalkyl substances with maternal early second trimester sex-steroid hormones. Int J Hyg Environ Health 2024; 259:114380. [PMID: 38657330 PMCID: PMC11127781 DOI: 10.1016/j.ijheh.2024.114380] [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: 10/12/2023] [Revised: 03/30/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND/AIMS Pregnant women are exposed to persistent environmental contaminants, including per- and polyfluoroalkyl substances (PFAS) that disrupt thyroid function. However, it is unclear if PFAS alter maternal sex-steroid hormone levels, which support pregnancy health and fetal development. METHODS In Illinois women with relatively high socioeconomic status (n = 460), we quantified perfluorononanoic (PFNA), perfluorooctane sulfonic (PFOS), perfluorooctanoic (PFOA), methyl-perfluorooctane sulfonamide acetic acid, perfluorohexanesulphonic (PFHxS), perfluorodecanoic (PFDeA), and perfluoroundecanoic (PFUdA) acid concentrations in fasting serum samples at median 17 weeks gestation, along with plasma progesterone, testosterone, and estradiol. We evaluated covariate-adjusted associations of ln-transformed hormones with each ln-transformed PFAS individually using linear regression and with the PFAS mixture using quantile-based g-computation (QGComp). RESULTS Interquartile range (IQR) increases in PFOS were associated with higher progesterone (%Δ 3.0; 95%CI: -0.6, 6.6) and estradiol (%Δ: 8.1; 95%CI: 2.2, 14.4) levels. Additionally, PFHxS was positively associated with testosterone (%Δ: 10.2; 95%CI: 4.0, 16.7), whereas both PFDeA and PFUdA were inversely associated with testosterone (%Δ: -5.7; 95%CI: -10.3, -0.8, and %Δ: -4.1; 95%CI: -7.6, -0.4, respectively). The IQR-standardized PFAS mixture was not associated with progesterone (%Δ: 1.6; 95%CI: -5.8, 9.2), due equal partial positive (%Δ: 9.2; driven by PFOA) and negative (%Δ: -7.4; driven by PFOS) mixture associations. Similarly, the mixture was not associated with testosterone (%Δ: 5.3; 95%CI: -9.0, 20.1), due to similar partial positive (%Δ: 23.6; driven by PFHxS) and negative (%Δ: -17.4; driven by PFDeA) mixture associations. However, we observed a slightly stronger partial positive (%Δ: 25.6; driven by PFOS and PFUdA) than negative (%Δ: -16.3; driven by PFOA) association resulting in an overall non-significant positive trend between the mixture and estradiol (%Δ: 8.5; 95%CI: -3.7, 20.9). CONCLUSION PFAS mixture modeled using QGComp was not associated with maternal sex-steroid hormones due to potential opposing effects of certain PFAS. Additional prospective studies could corroborate these findings.
Collapse
Affiliation(s)
- Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Libeth Rosas
- The Beckman Institute, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Jason Whalen
- Michigan Diabetes Research Center Chemistry Laboratory, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sabrina Smith
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA 94710, USA
| | - June-Soo Park
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA 94710, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94158, USA
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI 02912, USA
| | - Susan L Schantz
- The Beckman Institute, University of Illinois, Urbana-Champaign, IL 61801, USA; Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
9
|
Hull HR, Brown A, Gajewski B, Sullivan DK, Carlson SE. The Effect of Prenatal Docosahexaenoic Acid Supplementation on Offspring Fat Mass and Distribution at 24 Months Old. Curr Dev Nutr 2024; 8:103771. [PMID: 38948108 PMCID: PMC11214179 DOI: 10.1016/j.cdnut.2024.103771] [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: 03/04/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 07/02/2024] Open
Abstract
Background Excessive gestational weight gain (GWG) is related to increased offspring fat accrual, and increased fat mass (FM) is related to obesity development. Prenatal DHA supplementation has been linked to lower levels of offspring FM; however, conflicting data exist. Objectives This study aimed to determine if there is a protective effect of prenatal DHA supplementation on offspring fat accrual and adipose tissue deposition at 24 mo in offspring born to females who gain excessive weight compared with nonexcessive weight during pregnancy. We also explored if the effect of DHA dose on FM differed by offspring sex. Methods Infants born to females who participated in the Assessment of DHA on Reducing Early Preterm Birth randomized controlled trial (ADORE) were recruited. In ADORE, females were randomly assigned to either a high or low prenatal DHA supplement. Offspring body composition and adipose tissue distribution were measured using dual-energy x-ray absorptiometry (DXA). GWG was categorized as excessive or not excessive based on clinical guidelines. Results For total FM, there was a significant main effect for the DHA dose (P = 0.03); however, the dose by GWG status was nonsignificant (P = 0.44). Therefore, a higher prenatal DHA dose was related to greater offspring FM (622.9 g greater) and unrelated to GWG status. When investigating a DHA dose by sex effect, a significant main effect for DHA dose (P = 0.01) was detected for central FM. However, no interaction was detected (P = 0.98), meaning that both boys and girls had greater central FM if their mother was assigned to the higher DHA dose. Conclusions Greater prenatal DHA supplementation was associated with greater offspring FM and adipose tissue distribution at 24 mo. It will be important to understand if these effects persist into childhood.This trial was registered at clinicaltrials.gov as NCT03310983.
Collapse
Affiliation(s)
- Holly R Hull
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Alexandra Brown
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Byron Gajewski
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
10
|
Bonet ML, Ribot J, Sánchez J, Palou A, Picó C. Early Life Programming of Adipose Tissue Remodeling and Browning Capacity by Micronutrients and Bioactive Compounds as a Potential Anti-Obesity Strategy. Cells 2024; 13:870. [PMID: 38786092 PMCID: PMC11120104 DOI: 10.3390/cells13100870] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
The early stages of life, especially the period from conception to two years, are crucial for shaping metabolic health and the risk of obesity in adulthood. Adipose tissue (AT) plays a crucial role in regulating energy homeostasis and metabolism, and brown AT (BAT) and the browning of white AT (WAT) are promising targets for combating weight gain. Nutritional factors during prenatal and early postnatal stages can influence the development of AT, affecting the likelihood of obesity later on. This narrative review focuses on the nutritional programming of AT features. Research conducted across various animal models with diverse interventions has provided insights into the effects of specific compounds on AT development and function, influencing the development of crucial structures and neuroendocrine circuits responsible for energy balance. The hormone leptin has been identified as an essential nutrient during lactation for healthy metabolic programming against obesity development in adults. Studies have also highlighted that maternal supplementation with polyunsaturated fatty acids (PUFAs), vitamin A, nicotinamide riboside, and polyphenols during pregnancy and lactation, as well as offspring supplementation with myo-inositol, vitamin A, nicotinamide riboside, and resveratrol during the suckling period, can impact AT features and long-term health outcomes and help understand predisposition to obesity later in life.
Collapse
Affiliation(s)
- M. Luisa Bonet
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Joan Ribot
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (M.L.B.); (J.S.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
11
|
Hull HR, Gajewski BJ, Sullivan DK, Carson SE. Growth and adiposity in newborns study (GAINS): The influence of prenatal DHA supplementation protocol. Contemp Clin Trials 2023; 132:107279. [PMID: 37406769 PMCID: PMC10852997 DOI: 10.1016/j.cct.2023.107279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Obesity and central fat mass (FM) accrual drive disease development and are related to greater morbidity and mortality. Excessive gestational weight gain (GWG) increases fetal fat accretion resulting in greater offspring FM across the lifespan. Studies associate greater maternal docosahexaenoic acid (DHA) levels with lower offspring FM and lower visceral adipose tissue during childhood, however, most U.S. pregnant women do not consume an adequate amount of DHA. We will determine if prenatal DHA supplementation is protective for body composition changes during infancy and toddlerhood in offspring exposed to excessive GWG. METHODS AND DESIGN Infants born to women who participated in the Assessment of DHA on Reducing Early Preterm Birth randomized controlled trial (ADORE; NCT02626299) will be invited to participate. Women were randomized to either a high 1000 mg or low 200 mg daily prenatal DHA supplement starting in the first trimester of pregnancy. Offspring body composition and adipose tissue distribution will be measured at 2 weeks, 6 months, 12 months, and 24 months using dual energy x-ray absorptiometry. Maternal GWG will be categorized as excessive or not excessive based on clinical guidelines. DISCUSSION Effective strategies to prevent obesity development are lacking. Exposures during the prenatal period are important in the establishment of the offspring phenotype. However, it is largely unknown which exposures can be successfully targeted to have a meaningful impact. This study will determine if prenatal DHA supplementation modifies the relationship between maternal weight gain and offspring FM and FM distribution at 24 months of age. ETHICS AND DISSEMINATION The University of Kansas Medical Center Institutional Review Board (IRB) approved the study protocol (STUDY00140895). The results of the trial will be disseminated at conferences and in peer reviewed publications. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT03310983.
Collapse
Affiliation(s)
- Holly R Hull
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America.
| | - Byron J Gajewski
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Susan E Carson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
12
|
Wang Q, Wang X. The Effects of a Low Linoleic Acid/α-Linolenic Acid Ratio on Lipid Metabolism and Endogenous Fatty Acid Distribution in Obese Mice. Int J Mol Sci 2023; 24:12117. [PMID: 37569494 PMCID: PMC10419107 DOI: 10.3390/ijms241512117] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
A reduced risk of obesity and metabolic syndrome has been observed in individuals with a low intake ratio of linoleic acid/α-linolenic acid (LA/ALA). However, the influence of a low ratio of LA/ALA intake on lipid metabolism and endogenous fatty acid distribution in obese patients remains elusive. In this investigation, 8-week-old C57BL/6J mice were randomly assigned to four groups: low-fat diet (LFD) as a control, high-fat diet (HFD), high-fat diet with a low LA/ALA ratio (HFD+H3L6), and high-fat diet with a high LA/ALA ratio (HFD+L3H6) for 16 weeks. Our results show that the HFD+H3L6 diet significantly decreased the liver index of HFD mice by 3.51%, as well as the levels of triacylglycerols (TGs) and low-density lipoprotein cholesterol (LDL-C) by 15.67% and 10.02%, respectively. Moreover, the HFD+H3L6 diet reduced the pro-inflammatory cytokines interleukin-6 (IL-6) level and aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio and elevated the level of superoxide dismutase (SOD) in the liver. The HFD+H3L6 diet also resulted in the downregulation of fatty acid synthetase (FAS) and sterol regulatory element binding proteins-1c (SREBP-1c) expression and the upregulation of peroxisome proliferator-activated receptor-α (PPAR-α) and acyl-CoA oxidase 1 (ACOX1) gene expression in the liver. The low LA/ALA ratio diet led to a notable increase in the levels of ALA and its downstream derivative docosahexaenoic acid (DHA) in the erythrocyte, liver, perienteric fat, epididymal fat, perirenal fat, spleen, brain, heart, and gastrocnemius, with a strong positive correlation. Conversely, the accumulation of LA in abdominal fat was more prominent, and a high LA/ALA ratio diet exacerbated the deposition effect of LA. In conclusion, the low LA/ALA ratio not only regulated endogenous fatty acid levels but also upregulated PPAR-α and ACOX1 and downregulated SREBP-1c and FAS gene expression levels, thus maintaining lipid homeostasis. Optimizing dietary fat intake is important in studying lipid nutrition. These research findings emphasize the significance of understanding and optimizing dietary fat intake.
Collapse
Affiliation(s)
| | - Xingguo Wang
- State Key Laboratory of Food Science and Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
13
|
Pacyga DC, Haggerty DK, Gennings C, Schantz SL, Strakovsky RS. Interrogating Components of 2 Diet Quality Indices in Pregnancy using a Supervised Statistical Mixtures Approach. Am J Clin Nutr 2023; 118:290-302. [PMID: 37201722 PMCID: PMC10375457 DOI: 10.1016/j.ajcnut.2023.05.020] [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: 11/03/2022] [Revised: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND The Healthy Eating Index (HEI)-2015 and Alternative Healthy Eating Index (AHEI)-2010 evaluate diet holistically in pregnancy. However, it remains unclear how individual index components interact to contribute to health. OBJECTIVES To evaluate associations of HEI-2015 and AHEI-2010 components with gestational length using traditional and novel statistical methods in a prospective cohort. METHODS Pregnant women completed a 3-mo food-frequency questionnaire (FFQ) at median 13 wk gestation to calculate the HEI-2015 or AHEI-2010. Covariate-adjusted linear regression models evaluated associations of HEI-2015 and AHEI-2010 total scores and individual components (one at a time and simultaneously adjusted) with gestational length. Covariate-adjusted weighted quantile sum regression models evaluated 1) associations of HEI-2015 or AHEI-2010 components as mixtures with gestational length and 2) contributions of components to these associations. RESULTS Each 10-point increase in HEI-2015 and AHEI-2010 total score was associated with 0.11 (95% CI: -0.05, 0.27) and 0.14 (95% CI: 0.00, 0.28) wk longer gestation, respectively. In individual or simultaneously adjusted HEI-2015 models, higher intakes of seafood/plant proteins, total protein foods, greens/beans, and saturated fats but lower intakes of added sugars and refined grains were associated with longer gestational length. For the AHEI-2010, higher intake of nuts/legumes and lower intake of sugar-sweetened beverages (SSBs)/fruit juice were associated with longer gestational length. Jointly, 10% increases in HEI-2015 or AHEI-2010 mixtures were associated with 0.17 (95% CI: 0.001, 0.34) and 0.18 (95% CI: 0.05, 0.30) wk longer gestational length, respectively. Seafood/plant protein, total protein foods, dairy, greens/beans, and added sugars were the largest contributors to the HEI-2015 mixture. Nuts/legumes, SSBs/fruit juice, sodium, and DHA/EPA were the largest contributors to the AHEI-2010 mixture. Associations were less precise but consistent in women with spontaneous labors. CONCLUSIONS Compared to traditional methods, associations of diet index mixtures with gestational length were more robust and identified unique contributors. Additional studies could consider interrogating these statistical approaches using other dietary indices and health outcomes.
Collapse
Affiliation(s)
- Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Diana K Haggerty
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan L Schantz
- The Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL, USA; The Beckman Institute, University of Illinois, Urbana-Champaign, IL, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
14
|
Gómez-Vilarrubla A, Mas-Parés B, Carreras-Badosa G, Jové M, Berdún R, Bonmatí-Santané A, de Zegher F, Ibañez L, López-Bermejo A, Bassols J. Placental AA/EPA Ratio Is Associated with Obesity Risk Parameters in the Offspring at 6 Years of Age. Int J Mol Sci 2023; 24:10087. [PMID: 37373236 DOI: 10.3390/ijms241210087] [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: 05/19/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
During pregnancy, maternal polyunsaturated fatty acids (PUFA) are transferred to the fetus through the placenta by specific FA transporters (FATP). A higher perinatal exposure to n-6 over n-3 PUFA could be linked to excess fat mass and obesity development later in life. In this context, we aimed to assess the associations between long chain PUFAs (LC-PUFAs) (n-6, n-3, and n-6/n-3 ratios) measured in the placenta at term birth with obesity-related parameters in the offspring at 6 years of age and assess whether these associations are dependent on the placental relative expression of fatty acid transporters. As results, the PUFAn-6/PUFAn-3 ratio was 4/1, which scaled up to 15/1 when considering only the arachidonic acid/eicosapentaenoic acid ratio (AA/EPA ratio). Positive associations between the AA/EPA ratio and offspring's obesity risk parameters were found with weight-SDS, BMI-SDS, percent fat mass-SDS, visceral fat, and HOMA-IR (r from 0.204 to 0.375; all p < 0.05). These associations were more noticeable in those subjects with higher expression of fatty acid transporters. Therefore, in conclusion, a higher placental AA/EPA ratio is positively associated with offspring's visceral adiposity and obesity risk parameters, which become more apparent in subjects with higher expressions of placental FATPs. Our results support the potential role of n-6 and n-3 LC-PUFA in the fetal programming of obesity risk in childhood. For the present study, 113 healthy pregnant women were recruited during the first trimester of pregnancy and their offspring were followed up at 6 years of age. The fatty acid profiles and the expression of fatty acid transporters (FATP1 and FATP4) were analyzed from placental samples at birth. Associations between LC-PUFA (n-6, n-3, and n-6/n-3 ratios) and obesity risk parameters (weight, body mass index (BMI), percent fat mass, visceral fat, and homeostatic model assessment of insulin resistance (HOMA-IR)) in the offspring at 6 years of age were examined.
Collapse
Affiliation(s)
- Ariadna Gómez-Vilarrubla
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Berta Mas-Parés
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | - Rebeca Berdún
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | | | - Francis de Zegher
- Department of Development & Regeneration, University of Leuven, 3000 Leuven, Belgium
| | - Lourdes Ibañez
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children's Hospital, 08950 Esplugues de Llobregat, Spain
- CIBERDEM (Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders), ISCIII, 28029 Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
- Department of Pediatrics, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Department of Medical Sciences, University of Girona, 17003 Girona, Spain
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| |
Collapse
|
15
|
Francis EC, Kechris K, Jansson T, Dabelea D, Perng W. Novel Metabolic Subtypes in Pregnant Women and Risk of Early Childhood Obesity in Offspring. JAMA Netw Open 2023; 6:e237030. [PMID: 37014638 PMCID: PMC10074224 DOI: 10.1001/jamanetworkopen.2023.7030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 04/05/2023] Open
Abstract
Importance The in utero metabolic milieu is associated with offspring adiposity. Standard definitions of maternal obesity (according to prepregnancy body mass index [BMI]) and gestational diabetes (GDM) may not be adequate to capture subtle yet important differences in the intrauterine environment that could be involved in programming. Objectives To identify maternal metabolic subgroups during pregnancy and to examine associations of subgroup classification with adiposity traits in their children. Design, Setting, and Participants This cohort study included mother-offspring pairs in the Healthy Start prebirth cohort (enrollment: 2010-2014) recruited from University of Colorado Hospital obstetrics clinics in Aurora, Colorado. Follow-up of women and children is ongoing. Data were analyzed from March to December 2022. Exposures Metabolic subtypes of pregnant women ascertained by applying k-means clustering on 7 biomarkers and 2 biomarker indices measured at approximately 17 gestational weeks: glucose, insulin, Homeostatic Model Assessment for Insulin Resistance, total cholesterol, high-density lipoprotein cholesterol (HDL-C), triglycerides, free fatty acids (FFA), HDL-C:triglycerides ratio, and tumor necrosis factor α. Main Outcomes and Measures Offspring birthweight z score and neonatal fat mass percentage (FM%). In childhood at approximately 5 years of age, offspring BMI percentile, FM%, BMI in the 95th percentile or higher, and FM% in the 95th percentile or higher. Results A total of 1325 pregnant women (mean [SD] age, 27.8 [6.2 years]; 322 [24.3%] Hispanic, 207 non-Hispanic Black [15.6%], and 713 [53.8%] non-Hispanic White), and 727 offspring with anthropometric data measured in childhood (mean [SD] age 4.81 [0.72] years, 48% female) were included. We identified the following 5 maternal metabolic subgroups: reference (438 participants), high HDL-C (355 participants), dyslipidemic-high triglycerides (182 participants), dyslipidemic-high FFA (234 participants), and insulin resistant (IR)-hyperglycemic (116 participants). Compared with the reference subgroup, women in the IR-hyperglycemic and dyslipidemic-high FFA subgroups had offspring with 4.27% (95% CI, 1.94-6.59) and 1.96% (95% CI, 0.45-3.47) greater FM% during childhood, respectively. There was a higher risk of high FM% among offspring of the IR-hyperglycemic (relative risk, 8.7; 95% CI, 2.7-27.8) and dyslipidemic-high FFA (relative risk, 3.4; 95% CI, 1.0-11.3) subgroups; this risk was of greater magnitude compared with prepregnancy obesity alone, GDM alone, or both conditions. Conclusions and Relevance In this cohort study, an unsupervised clustering approach revealed distinct metabolic subgroups of pregnant women. These subgroups exhibited differences in risk of offspring adiposity in early childhood. Such approaches have the potential to refine understanding of the in utero metabolic milieu, with utility for capturing variation in sociocultural, anthropometric, and biochemical risk factors for offspring adiposity.
Collapse
Affiliation(s)
- Ellen C. Francis
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
| | - Katerina Kechris
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora
| | - Wei Perng
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora
| |
Collapse
|
16
|
Saros L, Vahlberg T, Koivuniemi E, Houttu N, Niinikoski H, Tertti K, Laitinen K. Fish Oil And/Or Probiotics Intervention in Overweight/Obese Pregnant Women and Overweight Risk in 24-Month-Old Children. J Pediatr Gastroenterol Nutr 2023; 76:218-226. [PMID: 36705702 PMCID: PMC9848211 DOI: 10.1097/mpg.0000000000003659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 01/28/2023]
Abstract
OBJECTIVES To evaluate whether a fish oil and/or probiotics intervention in pregnant women with overweight or obesity would influence the tendency of their 24-month-old children to become overweight and alter their body fat percentage. METHODS Women (n = 439) were double-blindly randomized into 4 intervention groups: fish oil+placebo, probiotics+placebo, probiotics+fish oil, and placebo+placebo (fish oil: 1.9 g docosahexaenoic acid and 0.22 g eicosapentaenoic acid, probiotics: Lacticaseibacillus rhamnosus HN001 and Bifidobacterium animalis ssp. lactis 420, 1010 colony-forming units each). The intervention lasted from early pregnancy until 6 months postpartum. Children's (n = 330) growth data (height, weight, head circumference), a secondary outcome of the trial, were evaluated at birth, 3, 6, 12, and 24 months of age and compared to Finnish growth charts. Body fat percentage was measured with air displacement plethysmography (24 months). Logistic regression and general linear models were used to analyze the data. RESULTS Probiotics+placebo [weight-for-height% adj. Odds ratio (OR) = 0.36, 95% confidence interval (CI) = 0.14-0.95] and probiotics+fish oil [weight-for-age standard deviation score (SD-score) adj. OR = 0.22, 95% CI = 0.07-0.71] associated with lower overweight odds in 24-month-old children compared to placebo+placebo. Results remained essentially the same, when probiotics' main effect (combined probiotics+placebo and probiotics+fish oil) was estimated; that is, lower overweight odds (weight-for-height% adj. OR = 0.48, 95% CI = 0.25-0.95 and weight-for-age SD-score adj. OR = 0.42, 95% CI = 0.20-0.88) compared to non-probiotics. No fish oil main effect (combined fish oil+placebo and probiotics+fish oil) was seen. The intervention did not influence body fat percentage. CONCLUSIONS The administration of probiotics solely and in combination with fish oil during pregnancy to women with overweight or obesity lowered the overweight odds of their 24-month-old children.
Collapse
Affiliation(s)
- Lotta Saros
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Tero Vahlberg
- the Institute of Clinical Medicine and Biostatistics, University of Turku, Turku, Finland
| | - Ella Koivuniemi
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Noora Houttu
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Harri Niinikoski
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- the Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Kristiina Tertti
- the Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, Turku, Finland
| | - Kirsi Laitinen
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Functional Foods Forum, University of Turku, Turku, Finland
| |
Collapse
|
17
|
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
|
18
|
Bahreynian M, Feizi A, Daniali SS, Kelishadi R. Interaction between maternal dietary fat intake, breast milk omega-3 fatty acids and infant growth during the first year of life. Child Care Health Dev 2023; 49:137-144. [PMID: 35751393 DOI: 10.1111/cch.13026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/05/2022] [Accepted: 06/19/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Long chain fatty acids (LCFA) of human milk (HM) might be associated with different patterns of weight gain in infancy. This study aims to examine the interaction between maternal dietary fat intake, breast milk content of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) with infant growth during the first year of life. METHODS This longitudinal study was conducted among 215 Iranian mother-infant pairs. The infants were followed up from birth to 12 months of age. Trained healthcare providers measured infant anthropometrics including weight, length and head circumference at birth, 2, 4, 6 and 12 months of age. Breast hind milk samples were collected in the morning through hand expression between 6 and 12 weeks postpartum. Maternal dietary intake was assessed using a 3-day dietary record. Linear mixed effects models were performed to determine the interaction between maternal dietary fat intake, the breast milk content of EPA and DHA with infant growth. Further adjustments were applied for potential confounders. RESULTS Mean and standard deviation (SD) of maternal age and body mass index were 29.70 (5.24) years and 24.47 (4.42) kg/m2 , respectively. Mean (SD) of infant birth weight was 3177.50 (413.20) g, and 50% were boys. An inverse interaction existed between maternal dietary fat intake and breast milk EPA with infant 4-month weight (β: -366.2, P-value: 0.01). We found a significant association of maternal dietary fat intake and infant 6-month weight (β: 22.5, P-value: 0.04). An inverse interaction was documented between maternal dietary fat intake and breast milk DHA with infant weight at 12-month of age (β: -95.3, P-value: 0.02). We found a significant association between maternal dietary fat intake and infant length at 12-month (β: 0.06, P-value: 0.02). CONCLUSION We found an inverse interaction between maternal dietary fat intake and breast milk omega-3 fatty acids with infant weight at 4 and 12 months of age. Although longer follow-up of growth indices is recommended, these findings suggest functional relevance of HM composition to infant growth.
Collapse
Affiliation(s)
- Maryam Bahreynian
- Department of Nutrition, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.,Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Biostatistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyede Shahrbanoo Daniali
- Department of Pediatrics, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Kelishadi
- Department of Pediatrics, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Infant Red Blood Cell Arachidonic to Docosahexaenoic Acid Ratio Inversely Associates with Fat-Free Mass Independent of Breastfeeding Exclusivity. Nutrients 2022; 14:nu14204238. [PMID: 36296922 PMCID: PMC9608835 DOI: 10.3390/nu14204238] [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: 09/14/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
The prevalence of childhood obesity has increased nearly ten times over the last 40 years, influenced by early life nutrients that have persistent effects on life-long metabolism. During the first six months, infants undergo accelerated adipose accumulation, but little is known regarding infant fatty acid status and its relationship to infant body composition. We tested the hypothesis that a low arachidonic to docosahexaenoic acid ratio (AA/DHA) in infant red blood cells (RBCs), a long-term indicator of fatty acid intake, would associate with more infant fat-free mass (FFM) and/or less adipose accumulation over the first 4 months of life. The fatty acid and composition of breastmilk and infant RBCs, as well as the phospholipid composition of infant RBCs, were quantified using targeted and unbiased lipid mass spectrometry from infants predominantly breastfed or predominantly formula-fed. Regardless of feeding type, FFM accumulation was inversely associated with the infant’s RBC AA/DHA ratio (p = 0.029, R2 = 0.216). Infants in the lowest AA/DHA ratio tertile had significantly greater FFM when controlling for infant sex, adiposity at 2 weeks, and feeding type (p < 0.0001). Infant RBC phospholipid analyses revealed greater peroxisome-derived ether lipids in the low AA/DHA group, primarily within the phosphatidylethanolamines. Our findings support a role for a low AA/DHA ratio in promoting FFM accrual and identify peroxisomal activity as a target of DHA in the growing infant. Both FFM abundance and peroxisomal activity may be important determinants of infant metabolism during development.
Collapse
|
20
|
Wu S, Zhao F, He Y, He T, Duan S, Feng G, Chen Y, Wang X, Szeto IMY, Lin L, Cai L. Association between maternal erythrocyte polyunsaturated fatty acid levels during pregnancy and offspring weight status: A birth cohort study. Front Nutr 2022; 9:978679. [PMID: 36245520 PMCID: PMC9557224 DOI: 10.3389/fnut.2022.978679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Background The findings of the association between maternal polyunsaturated fatty acid (PUFA) levels during pregnancy and offspring weight status are controversial. Furthermore, few studies have focused on Asian populations or used erythrocyte membranes as biological markers. We aimed to examine the associations between maternal erythrocyte PUFA and offspring weight status within the first 2 years among the Chinese population. Materials and methods A total of 607 mother-child pairs were recruited from a birth cohort. Maternal erythrocyte n-3 and n-6 PUFA during pregnancy were measured by gas chromatography, and the ratio of PUFA was calculated. Weight- and body mass index (BMI)-for-age z (WAZ and BAZ) scores were calculated for offspring at 1, 3, 6, 8, 12, 18, and 24 months of age. The risk of overweight and obesity was defined by the WHO criterion. The Generalized Estimating Equation (GEE) model was carried out for repeated anthropometric data within 2 years of age. Results Maternal erythrocyte docosapentaenoic acid (DPA, n-3) was inversely associated with offspring BAZ score [tertile 2 vs. tertile 1, β: −0.18 (−0.29, −0.00)]. Higher maternal erythrocyte arachidonic acid (AA) was inversely associated with lower offspring WAZ and BAZ [tertile 3 vs. tertile 1, β: −0.18 (−0.35, −0.02), −0.22 (−0.38, −0.06), respectively]. Furthermore, higher maternal erythrocyte AA [tertile 3 vs. tertile 1, odds ratio [OR]: 0.52 (0.36, 0.75), ptrend < 0.001] and total n-6 PUFA [tertile 3 vs. tertile 1, OR: 0.56 (0.39, 0.81), ptrend = 0.002] were associated with decreased risk of overweight and obesity in offspring. Maternal erythrocyte n-6/n-3 PUFA and AA/eicosapentaenoic acid (EPA) ratios were not associated with offspring weight status. Conclusion Maternal erythrocyte PUFA might influence offspring weight status within 2 years of age in the Chinese population. Further Asian studies are still needed.
Collapse
Affiliation(s)
- Shengchi Wu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Feng Zhao
- Institute of Nutrition & Health, Qingdao University, Qingdao, China
| | - Yannan He
- Institute of Nutrition & Health, Qingdao University, Qingdao, China
| | - Tingchao He
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Sufang Duan
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Nutrition and Health Research Center, National Center of Technology Innovation for Dairy, Hohhot, China
| | - Gang Feng
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Yujing Chen
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xin Wang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ignatius Man-Yau Szeto
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
- Nutrition and Health Research Center, National Center of Technology Innovation for Dairy, Hohhot, China
| | - Lizi Lin
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Li Cai
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Li Cai,
| |
Collapse
|
21
|
Li LJ, Wu J, Chen Z, Weir NL, Tsai MY, Albert P, Zhang C. Plasma phospholipid polyunsaturated fatty acids composition in early pregnancy and fetal growth trajectories throughout pregnancy: Findings from the US fetal growth studies-singletons cohort. EBioMedicine 2022; 82:104180. [PMID: 35853297 PMCID: PMC9294651 DOI: 10.1016/j.ebiom.2022.104180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/26/2022] Open
Abstract
Background We aimed to investigate plasma phospholipid PUFA levels in early pregnancy and fetal growth trajectories throughout pregnancy. Methods Within the NICHD Fetal Growth Studies–Singleton Cohort, we enrolled 2,802 pregnant women at gestational weeks 8–13 and randomly assigned them to four ultrasonogram schedules to capture weekly fetal growth throughout pregnancy. Eleven plasma phospholipid PUFAs were measured at early pregnancy using blood samples collected from a subsample of 321 pregnant women. We modeled fetal growth trajectories across tertiles of PUFAs with cubic splines using linear mixed models after adjusting for major confounders. We then compared pairwise weekly fetal growth biometrics referencing the lowest tertile in each PUFA using the Wald test. Findings Among plasma n-3 PUFAs in early pregnancy, docosahexaenoic acid (DHA, 22:6n3) and alpha-linolenic acid (ALA, 18:3n3) showed positive associations with all fetal growth measurements. For instance, compared with the lowest tertile, the highest tertile of DHA had greater estimated fetal growth (EFW) and abdominal circumference (AC), starting at 13 weeks of gestation and throughout pregnancy (at gestational week 38: 3235.3 vs. 3089.0 g for EFW; 344.6 vs. 339.2 mm for AC). As for plasma n-6 PUFAs, some showed positive associations (e.g., linoleic acid [LA], 18:2n6) while others (e.g., docosatetraenoic acid [DTA], 22:4n6) showed inverse associations with fetal growth measures. Interpretation Our data suggested that higher plasma levels of DHA and ALA in the first trimester were associated with increased fetal size and weight throughout subsequent pregnancy. Funding National Institute of Child Health and Human Development intramural funding.
Collapse
Affiliation(s)
- Ling-Jun Li
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Global Centre for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jing Wu
- Glotech Inc., Bethesda, Maryland, USA
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Natalie L Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul Albert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cuilin Zhang
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Global Centre for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
22
|
Metelcová T, Zamrazilová H, Vaňková M, Hill M, Tvrzická E, Staňková B, Taxová Braunerová R, Hainer V, Kunešová M. The fatty acid composition of serum phospholipids in adolescents is associated with body composition in early adulthoods: an eight-year follow-up study. Physiol Res 2022; 71:349-356. [PMID: 35616037 DOI: 10.33549/physiolres.934880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The fatty acid composition is associated with obesity. Omega 3 polyunsaturated fatty acid (PUFA) could have a beneficial role in the prevention and treatment of many disorders, including cardiometabolic diseases. A cohort of 84 men and 131 women were examined in adolescence and after 8 years. Body weight (BW) and fat mass (FM) were measured. The composition of fatty acids (FAs) of serum phospholipids was assessed using gas chromatography. Statistics: PLS method. Aim: to determine the relationships between FAs in adolescence and FM (explanatory variable 1, EV1) and BW (explanatory variable 2, EV2) in adulthood. In the predictive models, a cluster of FAs in boys explained 47.2 % of EV1 and a cluster of 6 FAs in girls explained 32.3 % of EV1 measured in adulthood. FAs measured in adolescents explained 23.7 % of EV2 in early adults regardless of gender. A significant negative association was found between 18:1n-9c and EV1 in males and EV2 in both genders. We found a significant negative association between 18:2n-6 and 20:0 and both EV1 and EV2. In all analyses, we found a significant negative association of 20:1n-9 and 18:3n-3 with EV1-2 in both genders. A significant positive association was found in 20:3n-6 with EV1 and EV2 in males. 20:4n-6 was positively associated with EV1 in females and EV2 in both genders. A positive association between FM and very long chain n- 6 PUFAs was also observed. It is concluded that serum MUFAs and essential PUFAs in adolescence are associated with lower BW and FM in adulthood.
Collapse
Affiliation(s)
- T Metelcová
- Institute of Endocrinology, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lin LH, Lin J, Yan JY. Interactive Affection of Pre-Pregnancy Overweight or Obesity, Excessive Gestational Weight Gain and Glucose Tolerance Test Characteristics on Adverse Pregnancy Outcomes Among Women With Gestational Diabetes Mellitus. Front Endocrinol (Lausanne) 2022; 13:942271. [PMID: 35872998 PMCID: PMC9301308 DOI: 10.3389/fendo.2022.942271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/07/2022] [Indexed: 12/16/2022] Open
Abstract
Purpose To examine the combined effect of pre-pregnancy overweight or obesity, excessive gestational weight gain, and glucose tolerance status on the incidence of adverse pregnancy outcomes among women with gestational diabetes mellitus. Methods A observational study including 5529 gestational diabetes mellitus patients was performed. Logistic regression were used to assess the independent and multiplicative interactions of overweight or obese, excessive gestational weight gain, abnormal items of oral glucose tolerance test and adverse pregnancy outcomes. Additive interactions were calculated using an Excel sheet developed by Anderson to calculate relative excess risk. Results Overall 1076(19.46%) study subject were overweight or obese and 1858(33.60%) women gained weight above recommended. Based on IADPSG criteria, more than one-third women with two, or three abnormal glucose values. Preconception overweight or obesity, above recommended gestational weight gain, and two or more abnormal items of oral glucose tolerance test parameters significantly increased the risk of adverse pregnancy outcomes, separately. After accounting for confounders, each two of overweight or obesity, excessive gestational weight gain, two or more abnormal items of OGTT parameters, the pairwise interactions on adverse pregnancy outcomes appear to be multiplicative. Coexistence of preconception overweight or obesity, above recommended gestational weight gain and two or more abnormal items of oral glucose tolerance test parameters increased the highest risk for adverse pregnancy outcomes. No additive interaction was found. Conclusions Pre-pregnancy overweight or obesity, excessive gestational weight gain, two or more abnormal items of OGTT parameters contribute to adverse pregnancy outcomes independently among women with gestational diabetes mellitus. Additionally, the combined effect between these three factors and adverse pregnancy outcomes appear to be multiplicative. Interventions focus on maternal overweight or obesity and gestational weight gain should be offered to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Li-hua Lin
- Department of Healthcare, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Juan Lin
- Department of Obstetrics, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Jian-ying Yan
- Department of Obstetrics, Fujian Maternity and Child Health Hospital, Fuzhou, China
| |
Collapse
|
24
|
Maternal Aerobic Exercise, but Not Blood Docosahexaenoic Acid and Eicosapentaenoic Acid Concentrations, during Pregnancy Influence Infant Body Composition. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148293. [PMID: 35886147 PMCID: PMC9316153 DOI: 10.3390/ijerph19148293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Although discrete maternal exercise and polyunsaturated fatty acid (PUFA) supplementation individually are beneficial for infant body composition, the effects of exercise and PUFA during pregnancy on infant body composition have not been studied. This study evaluated the body composition of infants born to women participating in a randomized control exercise intervention study. Participants were randomized to aerobic exercise (n = 25) or control (stretching and breathing) groups (n = 10). From 16 weeks of gestation until delivery, the groups met 3×/week. At 16 and 36 weeks of gestation, maternal blood was collected and analyzed for Docosahexaenoic Acid (DHA) and Eicosapentaenoic Acid (EPA). At 1 month postnatal, infant body composition was assessed via skinfolds (SFs) and circumferences. Data from 35 pregnant women and infants were analyzed via t-tests, correlations, and regression. In a per protocol analysis, infants born to aerobic exercisers exhibited lower SF thicknesses of triceps (p = 0.008), subscapular (p = 0.04), SF sum (p = 0.01), and body fat (BF) percentage (%) (p = 0.006) compared with controls. After controlling for 36-week DHA and EPA levels, exercise dose was determined to be a negative predictor for infant skinfolds of triceps (p = 0.001, r2 = 0.27), subscapular (p = 0.008, r2 = 0.19), SF sum (p = 0.001, r2 = 0.28), mid-upper arm circumference (p = 0.049, r2 = 0.11), and BF% (p = 0.001, r2 = 0.32). There were no significant findings for PUFAs and infant measures: during pregnancy, exercise dose, but not blood DHA or EPA levels, reduces infant adiposity.
Collapse
|
25
|
Associations of Cord Blood Lipids with Childhood Adiposity at the Age of Three Years: A Prospective Birth Cohort Study. Metabolites 2022; 12:metabo12060522. [PMID: 35736455 PMCID: PMC9231066 DOI: 10.3390/metabo12060522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/27/2022] Open
Abstract
We aimed to examine the associations between cord blood lipids and childhood adiposity and to investigate whether these associations vary across birth weight categories (small for gestational age (SGA), appropriate for gestational age (AGA) and large for gestational age (LGA)) in 1306 infants in the Born in Guangzhou Cohort Study, China. Adiposity outcomes at the age of three years included z-scores of weight-for-length/height (WFLZ), body mass index (BMIZ), subscapular (SSTZ) and triceps skinfold thickness (TSTZ), and the sum of skinfold thicknesses (SSFTZ). Cord blood triglycerides (TG) levels were negatively associated with WFLZ and BMIZ, whereas high density lipoprotein (HDL) levels were positively associated with WFLZ, BMIZ, TSTZ and SSFTZ. These associations were attenuated after adjustment for birth weight. Stratified analyses revealed that total cholesterol (TC) and low-density lipoprotein (LDL) levels were positively associated with childhood adiposity indicators among AGA infants but tended to be negatively associated with the adiposity indicators among LGA infants (p values for interaction <0.05). Furthermore, TG levels appeared to be positively associated with adiposity indicators among SGA infants but negatively associated with the outcomes among LGA infants (p values for interaction <0.05). Cord blood lipids levels might be associated with childhood adiposity, and these associations appear to differ across different birth weight categories. If confirmed in future studies, our findings suggest that individualized management plans might be warranted in preventing obesity.
Collapse
|
26
|
Yu HT, Xu WH, Chen YR, Ji Y, Tang YW, Li YT, Gong JY, Chen YF, Liu GL, Xie L. Association of Prepregnancy Obesity and Remodeled Maternal-Fetal Plasma Fatty Acid Profiles. Front Nutr 2022; 9:897059. [PMID: 35651505 PMCID: PMC9149296 DOI: 10.3389/fnut.2022.897059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Fatty acids, especially polyunsaturated fatty acid (PUFA), are found abundantly in the brain and are fundamental for a fetus's growth. The fatty acid profiles of mothers and fetuses may be affected by maternal prepregnancy body mass index (pre-BMI), thus affecting fetal growth and development. Methods A total of 103 mother-fetus pairs were divided into overweight/obese (OW, n = 26), normal weight (NW, n = 60), and underweight (UW, n = 17) groups according to pre-BMI. Fatty acid profiles in maternal and umbilical cord plasma were analyzed by gas chromatography. Results The infant birth BMI z-score of the OW group was higher than that of the NW and UW groups (p < 0.05). The OW mothers had significantly higher plasma n-6 PUFA and n-6/n-3, but lower docosahexaenoic acid (DHA) and n-3 PUFA (p < 0.05). In cord plasma, the proportions of DHA and n-3 PUFA were lower in the OW group (p < 0.05), whereas the n-6/n-3 ratio was higher in the OW group (p < 0.05). The pre-BMI was negatively correlated with cord plasma DHA in all subjects (r = −0.303, p = 0.002), and the same negative correlation can be observed in the OW group (r = −0.561, p = 0.004), but not in the NW and UW groups (p > 0.05). The pre-BMI was positively correlated with cord plasma n-6/n-3 in all subjects (r = 0.325, p = 0.001), and the same positive correlation can be found in the OW group (r = 0.558, p = 0.004), but not in NW and UW groups (p > 0.05). Conclusions Maternal pre-BMI was associated with the maternal-fetal plasma fatty acid profiles, whereas the adverse fatty acid profiles are more noticeable in the prepregnancy OW mothers.
Collapse
Affiliation(s)
- Hai-Tao Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Wen-Hui Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Yi-Ru Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Ye Ji
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Yi-Wei Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Yue-Ting Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Jia-Yu Gong
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Yi-Fei Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Guo-Liang Liu
- Experimental Teaching Center for Preventive Medicine, School of Public Health, Jilin University, Changchun, China
| | - Lin Xie
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
27
|
Algaba-Chueca F, Maymó-Masip E, Ballesteros M, Guarque A, Majali-Martínez A, Freixes O, Amigó N, Fernández-Veledo S, Vendrell J, Megía A. Cord Blood Advanced Lipoprotein Testing Reveals an Interaction between Gestational Diabetes and Birth-Weight and Suggests a New Early Biomarker of Infant Obesity. Biomedicines 2022; 10:biomedicines10051033. [PMID: 35625770 PMCID: PMC9138640 DOI: 10.3390/biomedicines10051033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Abnormal lipid metabolism is associated with gestational diabetes mellitus (GDM) and is observed in neonates with abnormal fetal growth. However, the underlying specific changes in the lipoprotein profile remain poorly understood. Thus, in the present study we used a novel nuclear magnetic resonance (NMR)-based approach to profile the umbilical cord serum lipoproteins. Two-dimensional diffusion-ordered 1H-NMR spectroscopy showed that size, lipid content, number and concentration of particles within their subclasses were similar between offspring born to control (n = 74) and GDM (n = 62) mothers. Subsequent data stratification according to newborn birth-weight categories, i.e., small (n = 39), appropriate (n = 50) or large (n = 49) for gestational age (SGA, AGA and LGA, respectively), showed an interaction between GDM and birth-weight categories for intermediate-density lipoproteins (IDL)-cholesterol content and IDL- and low-density lipoproteins (LDL)-triglyceride content, and the number of medium very low-density lipoproteins (VLDL) and LDL particles specifically in AGA neonates. Moreover, in a 2-year follow-up study, we observed that small LDL particles were independently associated with offspring obesity at 2 years (n = 103). Collectively, our data demonstrate that GDM disturbs triglyceride and cholesterol lipoprotein content across birth-weight categories, with AGA neonates born to GDM mothers displaying a profile more similar to that of adults with dyslipidemia. Furthermore, an altered fetal lipoprotein pattern was associated with the development of obesity at 2 years.
Collapse
Affiliation(s)
- Francisco Algaba-Chueca
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
| | - Elsa Maymó-Masip
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Mónica Ballesteros
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
- Department of Obstetrics and Gynecology, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain
| | - Albert Guarque
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
- Department of Obstetrics and Gynecology, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain
| | | | - Olga Freixes
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
| | - Núria Amigó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
- Biosfer Teslab SL Plaça del Prim, 10 2on 5a, 43201 Reus, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
| | - Joan Vendrell
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
- Correspondence: (J.V.); (A.M.); Tel.: +34-977-29-58-00 (A.M.)
| | - Ana Megía
- Department of Endocrinology and Nutrition and Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili (IISPV), Dr. Mallafre Guasch, 4, 43005 Tarragona, Spain; (F.A.-C.); (E.M.-M.); (O.F.); (S.F.-V.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departament of Basic Medical Sciences and Department of Medicine and Surgery, Rovira i Virgili University, 43005 Tarragona, Spain; (M.B.); (A.G.)
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria;
- Correspondence: (J.V.); (A.M.); Tel.: +34-977-29-58-00 (A.M.)
| |
Collapse
|
28
|
Age- and Diet-Dependent Changes in Hepatic Lipidomic Profiles of Phospholipids in Male Mice: Age Acceleration in Cyp2b-Null Mice. J Lipids 2022; 2022:7122738. [PMID: 35391786 PMCID: PMC8983274 DOI: 10.1155/2022/7122738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/03/2022] [Indexed: 11/17/2022] Open
Abstract
Increases in traditional serum lipid profiles are associated with obesity, cancer, and cardiovascular disease. Recent lipidomic analysis has indicated changes in serum lipidome profiles, especially in regard to specific phosphatidylcholines, associated with obesity. However, little work has evaluated murine hepatic liver lipidomic profiles nor compared these profiles across age, high-fat diet, or specific genotypes, in this case the lack of hepatic Cyp2b enzymes. In this study, the effects of age (9 months old), high-fat diet (4.5 months old), and the loss of three primarily hepatic xeno- and endobiotic metabolizing cytochrome P450 (Cyp) enzymes, Cyp2b9, Cyp2b10, and Cyp2b13 (Cyp2b-null mice), on the male murine hepatic lipidome were compared. Hierarchical clustering and principal component analysis show that age perturbs hepatic phospholipid profiles and serum lipid markers the most compared to young mice, followed by a high-fat diet and then loss of Cyp2b. Several lipid biomarkers such as PC/PE ratios, PE 38 : 6, and LPC concentrations indicate greater potential for NAFLD and hypertension with mixed effects in Cyp2b-null mice(less NAFLD and greater hypertension-associated markers). Lipid profiles from older mice contain greater total and n-6 fatty acids than normal diet (ND)-fed young mice; however, surprisingly, young Cyp2b-null mice contain high n-6 : n-3 ratios. Overall, the lack of Cyp2b typically enhanced adverse physiological parameters observed in the older (9 mo) mice with increased weight gain combined with a deteriorating cholesterol profile, but not necessarily all phospholipid profiles were adversely perturbed.
Collapse
|
29
|
Davis H, Magistrali A, Butler G, Stergiadis S. Nutritional Benefits from Fatty Acids in Organic and Grass-Fed Beef. Foods 2022; 11:foods11050646. [PMID: 35267281 PMCID: PMC8909876 DOI: 10.3390/foods11050646] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Livestock production is under increasing scrutiny as a component of the food supply chain with a large impact on greenhouse gas emissions. Amidst growing calls to reduce industrial ruminant production, there is room to consider differences in meat quality and nutritional benefits of organic and/or pasture-based management systems. Access to forage, whether fresh or conserved, is a key influencing factor for meat fatty acid profile, and there is increasing evidence that pasture access is particularly beneficial for meat’s nutritional quality. These composition differences ultimately impact nutrient supply to consumers of conventional, organic and grass-fed meat. For this review, predicted fatty acid supply from three consumption scenarios were modelled: i. average UK population National Diet and Nutrition Survey (NDNS) (<128 g/week) red meat consumption, ii. red meat consumption suggested by the UK National Health Service (NHS) (<490 g/week) and iii. red meat consumption suggested by the Eat Lancet Report (<98 g/week). The results indicate average consumers would receive more of the beneficial fatty acids for human health (especially the essential omega-3, alpha-linolenic acid) from pasture-fed beef, produced either organically or conventionally.
Collapse
Affiliation(s)
- Hannah Davis
- School of Natural and Environmental Science, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK; (A.M.); (G.B.)
- Correspondence: (H.D.); (S.S.)
| | - Amelia Magistrali
- School of Natural and Environmental Science, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK; (A.M.); (G.B.)
| | - Gillian Butler
- School of Natural and Environmental Science, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK; (A.M.); (G.B.)
| | - Sokratis Stergiadis
- Department of Animal Sciences, School of Agriculture, Policy and Development, University of Reading, Reading RG6 6EU, UK
- Correspondence: (H.D.); (S.S.)
| |
Collapse
|
30
|
Crawford SA, Christifano DN, Kerling EH, Gajewski BJ, Valentine CJ, Gustafson KM, Mathis NB, Camargo JT, Gibbs HD, Sullivan DK, Sands SA, Carlson SE. Validation of an abbreviated food frequency questionnaire for estimating DHA intake of pregnant women in the United States. Prostaglandins Leukot Essent Fatty Acids 2022; 177:102398. [PMID: 35063884 PMCID: PMC8825687 DOI: 10.1016/j.plefa.2022.102398] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 02/08/2023]
Abstract
Docosahexaenoic acid (DHA) intake was estimated in pregnant women between 12- and 20-weeks' gestation using the National Cancer Institute's (NCI) Diet History Questionnaire-II (DHQ-II) and a 7-question screener designed to capture DHA intake (DHA Food Frequency Questionnaire, DHA-FFQ). Results from both methods were compared to red blood cell phospholipid DHA (RBC-DHA) weight percent of total fatty acids. DHA intake from the DHA-FFQ was more highly correlated with RBC-DHA (rs=0.528) than the DHQ-II (rs=0.352). Moreover, the DHA-FFQ allowed us to obtain reliable intake data from 1355 of 1400 participants. The DHQ-II provided reliable intake for only 847 of 1400, because many participants only partially completed it and it was not validated for Hispanic participants. Maternal age, parity, and socioeconomic status (SES) were also significant predictors of RBC-DHA. When included with estimated intake from the DHA-FFQ, the model accounted for 36% of the variation in RBC-DHA.
Collapse
Affiliation(s)
- S A Crawford
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - D N Christifano
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America; The University of Kansas Medical Center, Hoglund Biomedical Imaging Center, Kansas City, KS, United States of America
| | - E H Kerling
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - B J Gajewski
- The University of Kansas Medical Center, Department of Biostatistics & Data Science, Kansas City, KS, United States of America
| | - C J Valentine
- Banner University Medical Center, The University of Arizona, Department of Pediatrics, Tucson, AZ, United States of America
| | - K M Gustafson
- The University of Kansas Medical Center, Hoglund Biomedical Imaging Center, Kansas City, KS, United States of America
| | - N B Mathis
- The University of Kansas Medical Center, Hoglund Biomedical Imaging Center, Kansas City, KS, United States of America
| | - J T Camargo
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - H D Gibbs
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - D K Sullivan
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - S A Sands
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America
| | - S E Carlson
- The University of Kansas Medical Center, Department of Dietetics and Nutrition, Kansas City, KS, United States of America.
| |
Collapse
|
31
|
Maternal polyunsaturated fatty acid concentrations during pregnancy and childhood liver fat accumulation. Clin Nutr 2022; 41:847-854. [DOI: 10.1016/j.clnu.2022.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022]
|
32
|
Longitudinal Associations of Plasma Phospholipid Fatty Acids in Pregnancy with Neonatal Anthropometry: Results from the NICHD Fetal Growth Studies-Singleton Cohort. Nutrients 2022; 14:nu14030592. [PMID: 35276951 PMCID: PMC8840515 DOI: 10.3390/nu14030592] [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: 01/03/2022] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Despite increasing interest in the health effects of polyunsaturated FAs (PUFAs), their roles in fetal and neonatal growth remain understudied. Within the NICHD Fetal Growth Studies—Singleton Cohort, we prospectively investigated the associations of individual and subclasses of plasma phospholipid PUFAs at gestational weeks (GW) 10−14, 15−26, 23−31, and 33−39 with neonatal anthropometric measures as surrogates for fetal growth among 107 women with gestational diabetes mellitus (GDM) and 214 non-GDM controls. Multivariable weighted linear regression models estimated the associations between plasma phospholipid PUFAs and neonatal anthropometric measures. Adjusted beta coefficients for phospholipid docosahexaenoic acid (DHA) per standard deviation (SD) increase at GW 23−31 in association with birthweight z-score, neonatal length, and neonatal fat mass were 0.25 (95% CI: 0.08−0.41), 0.57 (0.11−1.03) cm, and 54.99 (23.57−86.42) g, respectively; all false discovery rates (FDRs) < 0.05. Estimated Δ5-desaturase activity per SD increase at GW 33−39 but not at other time points was positively associated with birthweight z-score: 0.29 (95% CI: 0.08−0.33); neonatal length: 0.61 (0.29−0.94) cm; and neonatal fat mass: 32.59 (8.21−56.96) g; all FDRs < 0.05. Longitudinal analysis showed consistent results. Our findings suggest that mid-to-late pregnancy presented as critical windows for primarily diet-derived DHA and Δ5-desaturase activity in relation to neonatal anthropometric measures.
Collapse
|
33
|
Garcia-Irigoyen O, Bovenga F, Piglionica M, Piccinin E, Cariello M, Arconzo M, Peres C, Corsetto PA, Rizzo AM, Ballanti M, Menghini R, Mingrone G, Lefebvre P, Staels B, Shirasawa T, Sabbà C, Villani G, Federici M, Moschetta A. Enterocyte superoxide dismutase 2 deletion drives obesity. iScience 2022; 25:103707. [PMID: 35036884 PMCID: PMC8753186 DOI: 10.1016/j.isci.2021.103707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/19/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
Compelling evidence support an involvement of oxidative stress and intestinal inflammation as early events in the predisposition and development of obesity and its related comorbidities. Here, we show that deficiency of the major mitochondrial antioxidant enzyme superoxide dismutase 2 (SOD2) in the gastrointestinal tract drives spontaneous obesity. Intestinal epithelium-specific Sod2 ablation in mice induced adiposity and inflammation via phospholipase A2 (PLA2) activation and increased release of omega-6 polyunsaturated fatty acid arachidonic acid. Remarkably, this obese phenotype was rescued when fed an essential fatty acid-deficient diet, which abrogates de novo biosynthesis of arachidonic acid. Data from clinical samples revealed that the negative correlation between intestinal Sod2 mRNA levels and obesity features appears to be conserved between mice and humans. Collectively, our findings suggest a role of intestinal Sod2 levels, PLA2 activity, and arachidonic acid in obesity presenting new potential targets of therapeutic interest in the context of this metabolic disorder.
Collapse
Affiliation(s)
- Oihane Garcia-Irigoyen
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Fabiola Bovenga
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marilidia Piglionica
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Elena Piccinin
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marica Cariello
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Maria Arconzo
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Claudia Peres
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Paola Antonia Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via D. Trentacoste 2, 20133 Milan, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via D. Trentacoste 2, 20133 Milan, Italy
| | - Marta Ballanti
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Diabetes and Nutritional Sciences, Hodgkin Building, Guy's Campus, King's College London, London, UK
| | - Philippe Lefebvre
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Takuji Shirasawa
- Department of Molecular Gerontology, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Carlo Sabbà
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Gaetano Villani
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Massimo Federici
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonio Moschetta
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy.,IRCCS Istituto Tumori "Giovanni Paolo II", Viale O. Flacco 65, 70124 Bari, Italy
| |
Collapse
|
34
|
Perng W, Oken E. Programming long-term health: Maternal and fetal nutritional and dietary needs. EARLY NUTRITION AND LONG-TERM HEALTH 2022:27-63. [DOI: 10.1016/b978-0-12-824389-3.00008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
35
|
Zambrano E, Rodríguez-González GL, Reyes-Castro LA, Bautista CJ, Castro-Rodríguez DC, Juárez-Pilares G, Ibáñez CA, Hernández-Rojas A, Nathanielsz PW, Montaño S, Arredondo A, Huang F, Bolaños-Jiménez F. DHA Supplementation of Obese Rats throughout Pregnancy and Lactation Modifies Milk Composition and Anxiety Behavior of Offspring. Nutrients 2021; 13:nu13124243. [PMID: 34959795 PMCID: PMC8706754 DOI: 10.3390/nu13124243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 01/07/2023] Open
Abstract
We investigated if supplementing obese mothers (MO) with docosahexaenoic acid (DHA) improves milk long-chain polyunsaturated fatty acid (LCPUFA) composition and offspring anxiety behavior. From weaning throughout pregnancy and lactation, female Wistar rats ate chow (C) or a high-fat diet (MO). One month before mating and through lactation, half the mothers received 400 mg DHA kg−1 d−1 orally (C+DHA or MO+DHA). Offspring ate C after weaning. Maternal weight, total body fat, milk hormones, and milk nutrient composition were determined. Pups’ milk nutrient intake was evaluated, and behavioral anxiety tests were conducted. MO exhibited increased weight and total fat, and higher milk corticosterone, leptin, linoleic, and arachidonic acid (AA) concentrations, and less DHA content. MO male and female offspring had higher ω-6/ ω-3 milk consumption ratios. In the elevated plus maze, female but not male MO offspring exhibited more anxiety. MO+DHA mothers exhibited lower weight, total fat, milk leptin, and AA concentrations, and enhanced milk DHA. MO+DHA offspring had a lower ω-6/ω-3 milk intake ratio and reduced anxiety vs. MO. DHA content was greater in C+DHA milk vs. C. Supplementing MO mothers with DHA improves milk composition, especially LCPUFA content and ω-6/ω-3 ratio reducing offspring anxiety in a sex-dependent manner.
Collapse
Affiliation(s)
- Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
- Correspondence: ; Tel.: +52-55-5487-0900 (ext. 2417)
| | - Guadalupe L. Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | - Luis A. Reyes-Castro
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | - Claudia J. Bautista
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | - Diana C. Castro-Rodríguez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
- CONACyT-Cátedras, Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Gimena Juárez-Pilares
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | - Carlos A. Ibáñez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | - Alejandra Hernández-Rojas
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (G.L.R.-G.); (L.A.R.-C.); (C.J.B.); (D.C.C.-R.); (G.J.-P.); (C.A.I.); (A.H.-R.)
| | | | - Sara Montaño
- Department of Animal Nutrition, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Armando Arredondo
- Center for Health Systems Research, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Fengyang Huang
- Laboratory of Pharmacology and Toxicology, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Francisco Bolaños-Jiménez
- INRAE, UMR1280 Physiologie des Adaptations Nutritionnelles, Université de Nantes, Nantes Atlantique Université, 44096 Nantes, France;
| |
Collapse
|
36
|
Kim M, Voy BH. Fighting Fat With Fat: n-3 Polyunsaturated Fatty Acids and Adipose Deposition in Broiler Chickens. Front Physiol 2021; 12:755317. [PMID: 34658934 PMCID: PMC8511411 DOI: 10.3389/fphys.2021.755317] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
Modern broiler chickens are incredibly efficient, but they accumulate more adipose tissue than is physiologically necessary due to inadvertent consequences of selection for rapid growth. Accumulation of excess adipose tissue wastes feed in birds raised for market, and it compromises well-being in broiler-breeders. Studies driven by the obesity epidemic in humans demonstrate that the fatty acid profile of the diet influences adipose tissue growth and metabolism in ways that can be manipulated to reduce fat accretion. Omega-3 polyunsaturated fatty acids (n-3 PUFA) can inhibit adipocyte differentiation, induce fatty acid oxidation, and enhance energy expenditure, all of which can counteract the accretion of excess adipose tissue. This mini-review summarizes efforts to counteract the tendency for fat accretion in broilers by enriching the diet in n-3 PUFA.
Collapse
Affiliation(s)
| | - Brynn H. Voy
- Department of Animal Science, The University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
37
|
Maternal Dietary Quality and Dietary Inflammation Associations with Offspring Growth, Placental Development, and DNA Methylation. Nutrients 2021; 13:nu13093130. [PMID: 34579008 PMCID: PMC8468062 DOI: 10.3390/nu13093130] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
The ‘Developmental Origins of Health and Diseases’ hypothesis posits that prenatal maternal diet influences offspring growth and later life health outcomes. Dietary assessment has focused on selected nutrients. However, this approach does not consider the complex interactions between foods and nutrients. To provide a more comprehensive approach to public health, dietary indices have been developed to assess dietary quality, dietary inflammation and risk factors for non-communicable diseases. Thus far, their use in the context of placental development is limited and associations with offspring outcomes have been inconsistent. Although epidemiological studies have focused on the role of maternal diet on foetal programming, the underlying mechanisms are still poorly understood. Some evidence suggests these associations may be driven by placental and epigenetic changes. In this narrative review, we examine the current literature regarding relationships between key validated diet quality scores (Dietary Inflammatory Index [DII], Mediterranean diet [MD], Healthy Eating Index [HEI], Alternative Healthy Eating Index [AHEI], Dietary Approaches to Stop Hypertension [DASH], Glycaemic Index [GI] and Glycaemic Load [GL]) in pregnancy and birth and long-term offspring outcomes. We summarise findings, discuss potential underlying placental and epigenetic mechanisms, in particular DNA methylation, and highlight the need for further research and public health strategies that incorporate diet quality and epigenetics.
Collapse
|
38
|
Maternal DHA Supplementation during Pregnancy and Lactation in the Rat Protects the Offspring against High-Calorie Diet-Induced Hepatic Steatosis. Nutrients 2021; 13:nu13093075. [PMID: 34578953 PMCID: PMC8468499 DOI: 10.3390/nu13093075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
Maternal supplementation during pregnancy with docosahexaenoic acid (DHA) is internationally recommended to avoid postpartum maternal depression in the mother and improve cognitive and neurological outcomes in the offspring. This study was aimed at determining whether this nutritional intervention, in the rat, protects the offspring against the development of obesity and its associated metabolic disorders. Pregnant Wistar rats received an extract of fish oil enriched in DHA or saline (SAL) as placebo by mouth from the beginning of gestation to the end of lactation. At weaning, pups were fed standard chow or a free-choice, high-fat, high-sugar (fc-HFHS) diet. Compared to animals fed standard chow, rats exposed to the fc-HFHS diet exhibited increased body weight, liver weight, body fat and leptin in serum independently of saline or DHA maternal supplementation. Nevertheless, maternal DHA supplementation prevented both the glucose intolerance and the rise in serum insulin resulting from consumption of the fc-HFHS diet. In addition, animals from the DHA-fc-HFHS diet group showed decreased hepatic triglyceride accumulation compared to SAL-fc-HFHS rats. The beneficial effects on glucose homeostasis declined with age in male rats. Yet, the preventive action against hepatic steatosis was still present in 6-month-old animals of both sexes and was associated with decreased hepatic expression of lipogenic genes. The results of the present work show that maternal DHA supplementation during pregnancy programs a healthy phenotype into the offspring that was protective against the deleterious effects of an obesogenic diet.
Collapse
|
39
|
Chiu Y, Fadadu RP, Gaskins AJ, Rifas‐Shiman SL, Laue HE, Moley KH, Hivert M, Baccarelli A, Oken E, Chavarro JE, Cardenas A. Dietary fat intake during early pregnancy is associated with cord blood DNA methylation at IGF2 and H19 genes in newborns. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2021; 62:388-398. [PMID: 34288135 PMCID: PMC8364885 DOI: 10.1002/em.22452] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 05/03/2023]
Abstract
Maternal fat intake during pregnancy affects fetal growth, but mechanisms underlying this relationship are unclear. We performed an exploratory study of the associations of fat consumption during pregnancy with cord blood DNA methylation of the insulin-like growth factor 2 (IGF2) and H19 genes. We used data from 96 uncomplicated full-term pregnancies of mothers of whom majority had normal body mass index (BMI) (66%) in Project Viva, a prospective pre-birth cohort. We assessed maternal diet with validated food frequency questionnaires during the first and second trimesters and measured DNA methylation in segments of the IGF2- and H19-differentially methylated regions (DMRs) by pyrosequencing DNA extracted from umbilical cord blood samples. Mean (SD) age was 32.8 (4.1) years and prepregnancy BMI was 24.0 (4.4) kg/m2 . Mean DNA methylation was 56.3% (3.9%) for IGF2-DMR and 44.6% (1.9%) for H19-DMR. Greater first trimester intake of omega-6 polyunsaturated fat (effect per 1% of calories at the expense of carbohydrates) was associated with lower DNA methylation of IGF2-DMR (-1.2%; 95% confidence interval [CI]: -2.2%, -0.2%) and higher DNA methylation at H19-DMR (0.8%; 95% CI: 0.3%, 1.3%). On the other hand, greater first trimester intake of omega-3 polyunsaturated fat was associated with lower DNA methylation of the H19-DMR (-4.3%; 95% CI: -7.9%, -0.8%). We did not find significant associations of IGF2 and H19 methylation with IGF2 cord blood levels. Our findings suggest that early prenatal fat intake (omega-3, omega-6, and saturated fatty acids) may influence DNA methylation at the IGF2 and H19 locus, which could impact fetal development and long-term health.
Collapse
Affiliation(s)
- Yu‐Han Chiu
- Department of EpidemiologyHarvard TH Chan School of Public HealthBostonMassachusettsUSA
| | - Raj P. Fadadu
- School of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Division of Environmental Health SciencesUniversity of California, Berkeley School of Public Health, BerkeleyBerkeleyCaliforniaUSA
| | - Audrey J. Gaskins
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
| | - Sheryl L. Rifas‐Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Hannah E. Laue
- Department of EpidemiologyGeisel School of Medicine at Dartmouth CollegeHanoverNew HampshireUSA
| | - Kelle H. Moley
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Marie‐France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
- Diabetes Unit, Massachusetts General HospitalBostonMassachusettsUSA
| | - Andrea Baccarelli
- Department of Environmental Health SciencesMailman School of Public Health, Columbia UniversityNew York CityNew YorkUSA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Jorge E. Chavarro
- Department of EpidemiologyHarvard TH Chan School of Public HealthBostonMassachusettsUSA
- Department of NutritionHarvard TH Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Andres Cardenas
- Division of Environmental Health SciencesUniversity of California, Berkeley School of Public Health, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
40
|
Zou R, El Marroun H, Voortman T, Hillegers M, White T, Tiemeier H. Maternal polyunsaturated fatty acids during pregnancy and offspring brain development in childhood. Am J Clin Nutr 2021; 114:124-133. [PMID: 33742211 DOI: 10.1093/ajcn/nqab049] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 02/09/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Emerging evidence suggests an association of maternal PUFA concentrations during pregnancy with child cognitive and neuropsychiatric outcomes such as intelligence and autistic traits. However, little is known about prenatal maternal PUFAs in relation to child brain development, which may underlie these associations. OBJECTIVES We aimed to investigate the association of maternal PUFA status during pregnancy with child brain morphology, including volumetric and white matter microstructure measures. METHODS This study was embedded in a prospective population-based study. In total, 1553 mother-child dyads of Dutch origin were included. Maternal plasma glycerophospholipid PUFAs were assessed in midpregnancy. Child brain morphologic outcomes, including total gray and white matter volumes, as well as white matter microstructure quantified by global fractional anisotropy and mean diffusivity, were measured using MRI (including diffusion tensor imaging) at age 9-11 y. RESULTS Maternal ω-3 (n-3) long-chain PUFA (LC-PUFA) concentrations during pregnancy had an inverted U-shaped relation with child total gray volume (linear term: β: 16.7; 95% CI: 2.0, 31.5; quadratic term: β: -1.1; 95% CI: -2.1, -0.07) and total white matter volume (linear term: β: 15.7; 95% CI: 3.6, 27.8; quadratic term: β: -1.0; 95% CI: -1.8, -0.16). Maternal gestational ω-6 LC-PUFA concentrations did not predict brain volumetric differences in children, albeit the linolenic acid concentration was inversely associated with child total white matter volume. Maternal PUFA status during pregnancy was not related to child white matter microstructure. CONCLUSIONS Sufficient maternal ω-3 PUFAs during pregnancy may be related to more optimal child brain development in the long term. In particular, exposure to lower ω-3 PUFA concentrations in fetal life was associated with less brain volume in childhood. Maternal ω-6 LC-PUFAs were not related to child brain morphology.
Collapse
Affiliation(s)
- Runyu Zou
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,The Generation R Study Group, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hanan El Marroun
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Manon Hillegers
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tonya White
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Social and Behavioral Sciences, T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| |
Collapse
|
41
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
42
|
Satokar VV, Cutfield WS, Cameron-Smith D, Albert BB. Omega-3 fats in pregnancy: could a targeted approach lead to better metabolic health for children? Nutr Rev 2021; 79:574-584. [PMID: 32974665 DOI: 10.1093/nutrit/nuaa071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The prevalence of childhood obesity is increasing worldwide, and the children of women who are obese during pregnancy are at greatest risk. This risk may be mediated by exaggeration of the normal insulin resistance of pregnancy. Omega-3 (n-3) fats are insulin sensitizing. Supplementation during pregnancy may reduce metabolic risk and adiposity in the children. Though results from animal studies are encouraging, completed clinical trials have not demonstrated this benefit. However, to our knowledge, previous studies have not targeted women who are overweight or obese while pregnant-the group at greatest risk for insulin resistance and most likely to benefit from n-3. In this narrative review, the importance of performing clinical trials restricted to women who are overweight or obese is discussed, as is the potential importance of n-3 dose, oil source and quality, and the timing of the intervention.
Collapse
Affiliation(s)
- Vidit V Satokar
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| | - David Cameron-Smith
- Liggins Institute, University of Auckland, Auckland, New Zealand.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Benjamin B Albert
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| |
Collapse
|
43
|
Meinilä J, Klemetti MM, Huvinen E, Engberg E, Andersson S, Stach-Lempinen B, Koivusalo S. Macronutrient intake during pregnancy in women with a history of obesity or gestational diabetes and offspring adiposity at 5 years of age. Int J Obes (Lond) 2021; 45:1030-1043. [PMID: 33558642 PMCID: PMC8081655 DOI: 10.1038/s41366-021-00762-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 12/08/2020] [Accepted: 01/20/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND/OBJECTIVES The impact of maternal macronutrient intake during pregnancy on offspring childhood adiposity is unclear. We assessed the associations between maternal macronutrient intake during and after pregnancy with offspring adiposity at 5 years of age. Additionally, we investigated whether gestational diabetes (GDM), BMI, or breastfeeding modified these associations. SUBJECTS/METHODS Altogether, 301 mother-child dyads with maternal prepregnancy BMI ≥ 30 and/or previous GDM participated in the Finnish Gestational Diabetes Prevention Study (RADIEL) and its 5 years follow-up. Macronutrient intakes (E%) were calculated from 3-day food records collected at 5-18 weeks' gestation, in the third trimester, and at 12 months and 5 years after pregnancy. Offspring body fat mass (BFM) and fat percentage (BF%) at 5 years were measured by bioimpedance. Statistical analyses were multivariate linear regression. RESULTS Mean (SD) prepregnancy BMI was 33(4) kg/m2. GDM was diagnosed in 47%. In normoglycemic women, higher first half of pregnancy n-3 PUFA intake was associated with lower offspring BFM (g) (ß -0.90; 95% CI -1.62, -0.18) and BF% (ß -3.45; 95% CI -6.17, -0.72). In women with GDM, higher first half of pregnancy n-3 PUFA intake was associated with higher offspring BFM (ß 0.94; 95% CI 0.14, 1.75) and BF% (ß 3.21; 95% CI 0.43, 5.99). Higher SFA intake in the third trimester and cumulative intake across pregnancy (mean of the first half and late pregnancy) was associated with higher BFM and BF% (across pregnancy: ß 0.12; 95% CI 0.03, 0.20 and ß 0.44; 95% CI 0.15, 0.73, respectively). Higher carbohydrate intake across pregnancy was associated with lower BFM (ß -0.044; 95% CI -0.086, -0.003), and borderline associated with BF% (ß -0.15; 95% CI -0.31, 0.00). CONCLUSIONS The macronutrient composition of maternal diet during pregnancy is associated with offspring BFM and BF% at 5 years. GDM modifies the association between prenatal n-3 PUFA intake and offspring anthropometrics.
Collapse
Affiliation(s)
- Jelena Meinilä
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland.
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Miira M Klemetti
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, South Karelia Central Hospital, Lappeenranta, Finland
- Department of Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Emilia Huvinen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Elina Engberg
- Folkhälsan Research Center, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sture Andersson
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Beata Stach-Lempinen
- Department of Obstetrics and Gynecology, South Karelia Central Hospital, Lappeenranta, Finland
| | - Saila Koivusalo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
44
|
Huhtala MS, Rönnemaa T, Pellonperä O, Tertti K. Cord serum metabolome and birth weight in patients with gestational diabetes treated with metformin, insulin, or diet alone. BMJ Open Diabetes Res Care 2021; 9:e002022. [PMID: 34059525 PMCID: PMC8169462 DOI: 10.1136/bmjdrc-2020-002022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/09/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Recent research has demonstrated the benefits of metformin treatment in gestational diabetes (GDM) on short-term pregnancy outcomes (including excessive fetal growth and pre-eclampsia), but its effects on fetal metabolism remain mostly unknown. Our aim was to study the effects of metformin treatment compared with insulin or diet on the cord serum metabolome and also to assess how these metabolites are related to birth weight (BW) in pregnancies complicated by GDM. RESEARCH DESIGN AND METHODS Cord serum samples were available from 113, 97, and 98 patients with GDM treated with diet, insulin, and metformin, respectively. A targeted metabolome was measured using nuclear magnetic resonance spectroscopy. The patients in the metformin and insulin groups had participated in a previous randomized trial (NCT01240785). RESULTS Cord serum alanine was elevated in the metformin group (0.53 mmol/L) compared with the insulin (0.45 mmol/L, p<0.001) and the diet groups (0.46 mmol/L, p<0.0001). All other measured metabolites were similar between the groups. The triglyceride (TG)-to-phosphoglyceride ratio, average very low-density lipoprotein particle diameter, docosahexaenoic acid, omega-3 fatty acids (FAs), and ratios of omega-3 and monounsaturated FA to total FA were inversely related to BW. The omega-6-to-total-FA and omega-6-to-omega-3-FA ratios were positively related to BW. Cholesterol in very large and large high-density lipoprotein (HDL) was positively (p<0.01) associated with BW when adjusted for maternal prepregnancy body mass index, gestational weight gain, glycated hemoglobin, and mode of delivery. CONCLUSIONS Metformin treatment in GDM leads to an increase in cord serum alanine. The possible long-term implications of elevated neonatal alanine in this context need to be evaluated in future studies. Although previous studies have shown that metformin increased maternal TG levels, the cord serum TG levels were not affected. Cord serum HDL cholesterol and several FA variables are related to the regulation of fetal growth in GDM. Moreover, these associations seem to be independent of maternal confounding factors. TRIAL REGISTRATION NUMBER NCT01240785.
Collapse
Affiliation(s)
- Mikael S Huhtala
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| | - Tapani Rönnemaa
- Medicine, University of Turku, Turku, Finland
- Medicine, TYKS Turku University Hospital, Turku, Finland
| | - Outi Pellonperä
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| | - Kristiina Tertti
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| |
Collapse
|
45
|
Verduci E, Calcaterra V, Di Profio E, Fiore G, Rey F, Magenes VC, Todisco CF, Carelli S, Zuccotti GV. Brown Adipose Tissue: New Challenges for Prevention of Childhood Obesity. A Narrative Review. Nutrients 2021; 13:1450. [PMID: 33923364 PMCID: PMC8145569 DOI: 10.3390/nu13051450] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric obesity remains a challenge in modern society. Recently, research has focused on the role of the brown adipose tissue (BAT) as a potential target of intervention. In this review, we revised preclinical and clinical works on factors that may promote BAT or browning of white adipose tissue (WAT) from fetal age to adolescence. Maternal lifestyle, type of breastfeeding and healthy microbiota can affect the thermogenic activity of BAT. Environmental factors such as exposure to cold or physical activity also play a role in promoting and activating BAT. Most of the evidence is preclinical, although in clinic there is some evidence on the role of omega-3 PUFAs (EPA and DHA) supplementation on BAT activation. Clinical studies are needed to dissect the early factors and their modulation to allow proper BAT development and functions and to prevent onset of childhood obesity.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Animal Sciences for Health, Animal Production and Food Safety, University of Milan, 20133 Milan, Italy
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Vittoria Carlotta Magenes
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Carolina Federica Todisco
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| |
Collapse
|
46
|
Monthé-Drèze C, Sen S, Hauguel-de Mouzon S, Catalano PM. Effect of Omega-3 Supplementation in Pregnant Women with Obesity on Newborn Body Composition, Growth and Length of Gestation: A Randomized Controlled Pilot Study. Nutrients 2021; 13:nu13020578. [PMID: 33572368 PMCID: PMC7916127 DOI: 10.3390/nu13020578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity, a state of chronic low-grade metabolic inflammation, is a growing health burden associated with offspring adiposity, abnormal fetal growth and prematurity, which are all linked to adverse offspring cardiometabolic health. Higher intake of anti-inflammatory omega-3 (n-3) polyunsaturated fatty acids (PUFA) in pregnancy has been associated with lower adiposity, higher birthweight and longer gestation. However, the effects of n-3 supplementation specifically in pregnant women with overweight and obesity (OWOB) have not been explored. We conducted a pilot double-blind randomized controlled trial of 72 pregnant women with first trimester body mass index (BMI) ≥ 25 kg/m2 to explore preliminary efficacy of n-3 supplementation. Participants were randomized to daily DHA plus EPA (2 g/d) or placebo (wheat germ oil) from 10-16 weeks gestation to delivery. Neonatal body composition, fetal growth and length of gestation were assessed. For the 48 dyads with outcome data, median (IQR) maternal BMI was 30.2 (28.2, 35.4) kg/m2. In sex-adjusted analyses, n-3 supplementation was associated with higher neonatal fat-free mass (β: 218 g; 95% CI 49, 387) but not with % body fat or fat mass. Birthweight for gestational age z-score (-0.17 ± 0.67 vs. -0.61 ± 0.61 SD unit, p = 0.02) was higher, and gestation longer (40 (38.5, 40.1) vs. 39 (38, 39.4) weeks, p = 0.02), in the treatment vs. placebo group. Supplementation with n-3 PUFA in women with OWOB led to higher lean mass accrual at birth as well as improved fetal growth and longer gestation. Larger well-powered trials of n-3 PUFA supplementation specifically in pregnant women with OWOB should be conducted to confirm these findings and explore the long-term impact on offspring obesity and cardiometabolic health.
Collapse
Affiliation(s)
- Carmen Monthé-Drèze
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA;
- School of Medicine, Harvard University, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-617-525-4139
| | - Sarbattama Sen
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA;
- School of Medicine, Harvard University, Boston, MA 02115, USA
| | | | - Patrick M. Catalano
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA;
| |
Collapse
|
47
|
Behrouz V, Yari Z. A review on differential effects of dietary fatty acids on weight, appetite and energy expenditure. Crit Rev Food Sci Nutr 2020; 62:2235-2249. [PMID: 33261509 DOI: 10.1080/10408398.2020.1852172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The association between weight and chronic diseases is well defined. The quality and quantity of dietary fatty acids is an important external factor and appetite and energy expenditure, are important internal factors in determining body weight. On the other hand, dietary fatty acids composition can modulate appetite and energy metabolism, but not all fats are equal in producing metabolic responses.Given the accumulating evidence for differential effects of various dietary fatty acids, one important area of investigation is to scrutinize their roles in weight, appetite and energy expenditure modulation. There is substantial evidence to suggest that saturated fatty acids have a greater effect on appetite control, although in the long run may result in more weight gain than unsaturated fatty acids due to a weaker stimulation of energy expenditure. In contrast, mono-unsaturated fats do not have much effects on appetite control, but they can be beneficial in weight control over the long term due to stimulatory effects on energy expenditure. Interestingly, in case of poly unsaturated fats, including n-3 and n-6, their effect on increasing energy expenditure is aligned, but they act differently in controlling weight and appetite.
Collapse
Affiliation(s)
- Vahideh Behrouz
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Yari
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Maternal Linoleic Acid Overconsumption Alters Offspring Gut and Adipose Tissue Homeostasis in Young but Not Older Adult Rats. Nutrients 2020; 12:nu12113451. [PMID: 33187208 PMCID: PMC7697261 DOI: 10.3390/nu12113451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/26/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal n-6 polyunsaturated fatty acids (PUFA) consumption during gestation and lactation can predispose offspring to the development of metabolic diseases such as obesity later in life. However, the mechanisms underlying the potential programming effect of n-6 PUFA upon offspring physiology are not yet all established. Herein, we investigated the effects of maternal and weaning linoleic acid (LA)-rich diet interactions on gut intestinal and adipose tissue physiology in young (3-month-old) and older (6-month-old) adult offspring. Pregnant rats were fed a control diet (2% LA) or an LA-rich diet (12% LA) during gestation and lactation. At weaning, offspring were either maintained on the maternal diet or fed the other diet for 3 or 6 months. At 3 months of age, the maternal LA-diet favored low-grade inflammation and greater adiposity, while at 6 months of age, offspring intestinal barrier function, adipose tissue physiology and hepatic conjugated linoleic acids were strongly influenced by the weaning diet. The maternal LA-diet impacted offspring cecal microbiota diversity and composition at 3 months of age, but had only few remnant effects upon cecal microbiota composition at 6 months of age. Our study suggests that perinatal exposure to high LA levels induces a differential metabolic response to weaning diet exposure in adult life. This programming effect of a maternal LA-diet may be related to the alteration of offspring gut microbiota.
Collapse
|
49
|
Ghassabian A, Hornig M, Chen Z, Yeung E, Buka SL, Yu J, Ma G, Goldstein JM, Gilman SE. Gestational Cytokines and the Developmental Expression of Obesity in Childhood. Obesity (Silver Spring) 2020; 28:2192-2200. [PMID: 32985128 PMCID: PMC7644634 DOI: 10.1002/oby.22967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study examined the extent to which maternal immune activity during pregnancy is associated with childhood adiposity, and if so, whether associations at birth differ from those in infancy and childhood. Sex-specific associations were also examined. METHODS Participants were 1,366 singleton pregnancies from the Collaborative Perinatal Project (1959-1966). Interleukin-1β (IL-1β), IL-6, TNF-α, IL-8, and IL-10 in maternal sera were assayed repeatedly during pregnancy. Children's BMI was calculated repeatedly from birth through age 8 and derived age- and sex-normalized BMI z scores (BMIz). Linear mixed models were used to estimate the cumulative concentration of each cytokine in the second and third trimesters and then related this concentration to child BMIz. RESULTS Children exposed to higher IL-1β, IL-6, IL-8, and IL-10 concentrations had lower BMIz at birth but higher BMIz during childhood. Higher concentrations of IL-8 and IL-1β were also associated with higher BMIz during infancy (B per log increase in IL-8 = 0.04; 95% CI: 0.02 to 0.07; B per log increase in IL-1β = 0.03; 95% CI: 0.001 to 0.06). The associations between TNF-α and BMIz were in opposing directions in boys (B = -0.13; 95% CI: -0.31 to 0.04) and girls (B = 0.14; 95% CI: 0.02 to 0.26) during childhood. CONCLUSIONS Maternal prenatal inflammation contributes to the age- and sex-specific programming of obesity risk in childhood.
Collapse
Affiliation(s)
- Akhgar Ghassabian
- Departments of Pediatrics, Environmental Medicine, and Population Health, New York University School of Medicine, New York, NY, USA
| | - Mady Hornig
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Zhen Chen
- Biostatistics and Bioinformatics Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Edwina Yeung
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Stephen L Buka
- Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island
| | - Jing Yu
- Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gina Ma
- Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Jill M Goldstein
- Departments of Psychiatry and Medicine, Harvard Medical School, and Department of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Stephen E Gilman
- Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
50
|
Flannagan K, Gahagan S, Das A, Burrows R, Lozoff B, Villamor E. Serum polyunsaturated fatty acids in infancy are associated with body composition in adolescence. Pediatr Obes 2020; 15:e12656. [PMID: 32426929 DOI: 10.1111/ijpo.12656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 04/07/2020] [Accepted: 04/23/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Polyunsaturated fatty acids (PUFA) have been related to the development of adiposity. N-3 PUFA appears to be protective against obesity risk, while n-6 PUFA may be associated with greater adiposity. However, most studies have been conducted among adults. The role of PUFA in infancy is unknown. OBJECTIVE To examine associations of serum PUFA at age 1 year with age- and sex-adjusted body mass index Z score (BMIZ) change through age 16 years and body composition at 16 years. METHODS We quantified serum PUFA in 636 Chilean infants aged 1 year. We measured BMIZ at ages 1, 5, 10 and 16 years, and body composition by dual energy X-ray absorptiometry at 16 years. We estimated differences in 1- to 16-years BMIZ change between PUFA quartiles from multivariable linear mixed models with restricted cubic splines. At 16 years, we estimated differences in total fat mass (ToFM), truncal fat mass (TrFM), total lean mass (TLM), percent total fat mass (%ToFM) and percent truncal fat mass (%TrFM) between PUFA quartiles using linear regression. RESULTS PUFA were not associated with BMIZ change. Alpha-linolenic acid (ALA) was positively associated with TrFM (P = .03) and %TrFM (P < .0001) at 16 years while eicosapentaenoic acid (EPA) was inversely associated with %TrFM (P = .001). Docosapentaenoic acid (DPA) was positively associated with ToFM (P = .01), TrFM (P = .009), %ToFM (P = .02) and %TrFM (P = .02). Gamma-linolenic acid (GLA) and the Δ6-desaturase (D6D) activity index were each positively, linearly associated with ToFM, TrFM and %ToFM. The Δ5-desaturase (D5D) activity index was inversely associated with %TrFM (P = .04). CONCLUSIONS ALA, DPA, GLA and the D6D index at 1 year of age were positively associated with adiposity at age 16 years, while EPA and the D5D index were inversely associated with central adiposity. Our results related to EPA and desaturase indices are in agreement with limited prior studies.
Collapse
Affiliation(s)
- Kerry Flannagan
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Sheila Gahagan
- Division of Child Development and Community Health, University of California, San Diego, California, USA
| | - Arun Das
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Raquel Burrows
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Betsy Lozoff
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Eduardo Villamor
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|