1
|
Vacy K, Rupasinghe T, Bjorksten A, Gogos A, Meikle PJ, Burugupalli S, Boon WC, Ponsonby AL. The associations between prenatal plastic phthalate exposure and lipid acylcarnitine levels in humans and mice. Reprod Toxicol 2025; 132:108835. [PMID: 39800279 DOI: 10.1016/j.reprotox.2025.108835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Phthalates are ubiquitous environmental pollutants known for their endocrine-disrupting properties, particularly during critical periods such as pregnancy and early childhood. Phthalates alter lipid metabolism, but the role of prenatal exposure on the offspring lipidome is less understood. In particular, we focused on long chain acylcarnitines - intermediates of fatty acid oxidation that serve as potential biomarkers of mitochondrial function and energy metabolism. This study aimed (i) to investigate the association between prenatal phthalate exposure and the child's blood acylcarnitine concentrations and, (ii) to evaluate the impact of prenatal administration of di-(2-ethylhexyl) phthalate (DEHP) on acylcarnitine levels in mouse offspring blood, brain and liver. We conducted analyses of both a prospective birth cohort study and an experimental study in mice. From the Barwon Infant Study cohort (1074 mother-child pairs), prenatal phthalate exposure was assessed at 36 weeks' gestation and its association with acylcarnitine levels was examined in cord blood, and child's blood at 6 months, 12 months and 4 years. In mice, pregnant C57BL/6 J mouse dams were exposed to 20 μg/kg DEHP for 5 days mid-gestation, and offspring tissues were analyzed at 1 month of age postnatally for acylcarnitine profiles. Our findings demonstrate that prenatal phthalate levels (specifically butyl benzyl phthalate (BBzP) and diisobutyl phthalate (DiBP)) are inversely associated with total long chain acylcarnitine levels in human cord blood at birth. In contrast, BBzP was positively associated with the long chain acylcarnitines at 12 months of age. In mice, prenatal DEHP exposure for only 5 days led to decreased palmitoylcarnitine (AC16:0) levels in the brain and liver, but not in blood. Taken together, our findings highlight that prenatal phthalate exposure can alter acylcarnitine profiles, indicating disruptions in fatty acid metabolism that may have long-term effects on metabolic health.
Collapse
Affiliation(s)
- Kristina Vacy
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville 3010, Australia
| | | | - Alicia Bjorksten
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora 3086, Australia
| | - Satvika Burugupalli
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora 3086, Australia
| | - Wah Chin Boon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
2
|
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
|
3
|
Liu S, Hua L, Mo X, Lei B, Zhang R, Zhou S, Jiang X, Fang Z, Feng B, Che L, Xu S, Lin Y, Wu D, Zhuo Y, Jin C. Comparative Impact of Alternate-Day Fasting and Time-Restricted Feeding on Placental Function and Fetal Development in Maternal Obesity. Nutrients 2024; 17:25. [PMID: 39796458 PMCID: PMC11723168 DOI: 10.3390/nu17010025] [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: 11/09/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Maternal obesity detrimentally affects placental function and fetal development. Both alternate-day fasting (ADF) and time-restricted feeding (TRF) are dietary interventions that can improve metabolic health, yet their comparative effects on placental function and fetal development remain unexplored. OBJECTIVES This study aims to investigate the effects of ADF and TRF on placental function and fetal development during maternal consumption of a high-fat diet (HFD). METHODS One hundred 8-week-old female mice were assigned to one of four dietary regimens: (1) normal diet with ad libitum feeding (NA); (2) HFD with ad libitum feeding (HA); (3) HFD with ADF (HI); and (4) HFD with TRF (HT), administered both before and during pregnancy. On gestational day 18.5, serum and placental samples were collected from both mothers and fetuses to examine placental function and fetal development. RESULTS During gestation, both ADF and TRF substantially alleviated the metabolic impairments caused by an HFD in obese maternal mice. TRF mice demonstrated enhanced placental nutrient transport and fetal development, associated with reduced endoplasmic reticulum (ER) stress and inflammatory responses. In contrast, ADF markedly intensified placental stress and inflammatory responses, diminished placental nutrient transport efficiency, and consequently induced fetal growth restriction. CONCLUSIONS Both ADF and TRF during pregnancy significantly mitigated metabolic impairments in obese dams on an HFD. TRF mice demonstrated enhanced placental nutrient transport and fetal development, associated with reduced endoplasmic reticulum (ER) stress and inflammatory responses. In contrast, ADF markedly intensified placental stress and inflammatory responses, diminished placental nutrient transport efficiency, and consequently induced fetal growth restriction.
Collapse
Affiliation(s)
- Siyuan Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Xi Mo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Bing Lei
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Ruihao Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Shihao Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Xuemei Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Chao Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| |
Collapse
|
4
|
Azuma D, Penner Y, Kaneko-Tarui T, Mahmoud T, Breeze JL, Rodday A, O’Tierney-Ginn P, Maron JL. Neonatal body composition, salivary feeding gene expression, and feeding outcomes in infants of diabetic mothers. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2024; 5:1501805. [PMID: 39749266 PMCID: PMC11693610 DOI: 10.3389/fcdhc.2024.1501805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Introduction Infants of diabetic mothers (IDMs) may exhibit decreased oral intake, requiring nasogastric feedings and prolonged hospitalization. The objective of this study was to explore whether saliva serves as an informative biofluid for detecting expression of hunger signaling and energy homeostasis modulator genes and to perform exploratory analyses examining expression profiles, body composition, and feeding outcomes in late preterm and term IDMs and infants born to mothers with normoglycemia during pregnancy. Methods In this prospective cohort pilot study, infants born at ≥ 35 weeks' gestation to mothers with gestational or type II diabetes (IDM cohort) and normoglycemic mothers (control cohort) were recruited. The presence of known hunger signaling genes: 5'AMP-activated protein kinase (PRKAA2) and neuropeptide Y2 receptor (NPY2R); adipokines: leptin (LEP) and adiponectin (ADIPOQ); and energy homeostasis regulators: ghrelin (GHRL) and proopiomelanocortin (POMC) in neonatal saliva was determined with RT-qPCR and compared between cohorts. Body composition was assessed via skinfold measurements and compared between cohorts. Feeding outcomes were recorded. Exploratory analyses were performed examining associations between infant body composition, energy homeostasis and hunger signaling gene expression. Results Twenty-three infants in the IDM cohort and 22 infants in the control cohort were recruited. LEP and ADIPOQ were not reliably detected in neonatal saliva in either cohort. PRKAA2, GHRL and NPY2R were less likely to be detected in the IDM cohort, whereas POMC was more likely to be detected in the IDM cohort. Infants in the IDM cohort had greater adiposity compared to infants in the normoglycemia cohort. Only 3 IDMs had documented poor feeding; no infant in the control group struggled to feed. In exploring associations between hunger signaling gene expression with energy homeostasis gene expression and body composition, the odds of detecting salivary NPY2R expression decreased as fat mass increased, and the odds of detecting PRKAA2 expression increased in the presence of GHRL expression. Discussion Non-invasive assessment of hunger signaling and energy homeostasis gene expression is possible through neonatal salivary analysis. This pilot study lays the foundation for a larger scale study to further investigate the link between in utero exposure to diabetes with body composition and regulation of appetite.
Collapse
Affiliation(s)
- Dara Azuma
- Mother Infant Research Institute at Tufts Medical Center, Boston, MA, United States
| | - Yvette Penner
- Frances Stern Nutrition Center at Tufts Medical Center, Boston, MA, United States
| | - Tomoko Kaneko-Tarui
- Mother Infant Research Institute at Tufts Medical Center, Boston, MA, United States
| | - Taysir Mahmoud
- Mother Infant Research Institute at Tufts Medical Center, Boston, MA, United States
| | - Janis L. Breeze
- Clinical and Translational Science Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA, United States
| | - Angie Rodday
- Clinical and Translational Science Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA, United States
| | | | - Jill L. Maron
- Mother Infant Research Institute at Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
5
|
Sahoo PK, Ravi A, Liu B, Yu J, Natarajan SK. Palmitoleate protects against lipopolysaccharide-induced inflammation and inflammasome activity. J Lipid Res 2024; 65:100672. [PMID: 39396700 PMCID: PMC11585775 DOI: 10.1016/j.jlr.2024.100672] [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: 02/06/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024] Open
Abstract
Inflammation is part of natural immune defense mechanism against any form of infection or injury. However, prolonged inflammation could perturb cell homeostasis and contribute to the development of metabolic and inflammatory diseases, including maternal obesity, diabetes, cardiovascular diseases, and metabolic dysfunction-associated steatotic liver diseases (MASLD). Polyunsaturated fatty acids have been shown to mitigate inflammatory response by generating specialized proresolving lipid mediators, which take part in resolution of inflammation. Similarly here, we show that palmitoleate, an omega-7 monounsaturated fatty acid exerts anti-inflammatory properties in response to lipopolysaccharide (LPS)-mediated inflammation. Exposure of bone marrow-derived macrophages (BMDMs) to LPS or TNFα induces robust increase in the expression of proinflammatory cytokines and supplementation of palmitoleate inhibited LPS-mediated upregulation of proinflammatory cytokines. We also observed that palmitoleate was able to block LPS + ATP-induced inflammasome activation mediated cleavage of procaspase 1 and prointerleukin-1β. Further, treatment of palmitoleate protects against LPS-induced inflammation in human THP-1-derived macrophages and trophoblasts. Coexposure of LPS and palmitate (saturated free fatty acid) induces inflammasome and cell death in BMDMs, however, treatment of palmitoleate blocked LPS and palmitate-induced cell death in BMDMs. Further, LPS and palmitate together results in the activation of mitogen-activated protein kinases and pretreatment of palmitoleate inhibited the activation of mitogen-activated protein kinases and nuclear translocation of nuclear factor kappa B in BMDMs. In conclusion, palmitoleate shows anti-inflammatory properties against LPS-induced inflammation and LPS + palmitate/ATP-induced inflammasome activity and cell death.
Collapse
Affiliation(s)
- Prakash Kumar Sahoo
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Aiswariya Ravi
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Baolong Liu
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yang ling, Shaanxi, China
| | - Jiujiu Yu
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; College of Allied Health Professions Medical Nutrition Education, University of Nebraska Medical Center, Omaha, NE, USA; Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
6
|
Fox A, Hayes B, Doyle E. Advancing our knowledge of placental function and how it relates to the developing fetal brain. Semin Fetal Neonatal Med 2024; 29:101549. [PMID: 39550256 DOI: 10.1016/j.siny.2024.101549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2024]
Abstract
Perinatal medicine has made significant advancements in recent decades. This has improved care and outcomes for infants. As we strive to improve neurodevelopmental outcomes, we must understand the influence the maternal/placental/fetal (MPF) triad has on fetal development and postnatal health and disease. Our understanding of the MPF triad remains incomplete, however research is continuing to develop our understanding. Through further research and incorporating what is currently known into how we deliver perinatal care, we have the opportunity to improve outcomes for infants. This review focuses on what is currently known about the structure and function of the placenta and the influence of the MPF triad. Current modalities for assessment of the MPF triad and future avenues for research will also be discussed. Understanding the relationship between the MPF triad, neurodevelopment and long-term health and disease has the potential to open new avenues for disease prevention and treatment through the lifespan.
Collapse
Affiliation(s)
- Aine Fox
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland; Royal College of Surgeons (RCSI), Dublin, Ireland.
| | - Breda Hayes
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland; Royal College of Surgeons (RCSI), Dublin, Ireland.
| | - Emma Doyle
- Department of Histopathology, Rotunda Hospital, Dublin, Ireland.
| |
Collapse
|
7
|
Grismaldo R A, Luévano-Martínez LA, Reyes M, García-Márquez G, García-Rivas G, Sobrevia L. Placental mitochondrial impairment and its association with maternal metabolic dysfunction. J Physiol 2024. [PMID: 39116002 DOI: 10.1113/jp285935] [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: 03/23/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
The placenta plays an essential role in pregnancy, leading to proper fetal development and growth. As an organ with multiple physiological functions for both mother and fetus, it is a highly energetic and metabolically demanding tissue. Mitochondrial physiology plays a crucial role in the metabolism of this organ and thus any alteration leading to mitochondrial dysfunction has a severe outcome in the development of the fetus. Pregnancy-related pathological states with a mitochondrial dysfunction outcome include preeclampsia and gestational diabetes mellitus. In this review, we address the role of mitochondrial morphology, metabolism and physiology of the placenta during pregnancy, highlighting the roles of the cytotrophoblast and syncytiotrophoblast. We also describe the relationship between preeclampsia, gestational diabetes, gestational diabesity and pre-pregnancy maternal obesity with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Adriana Grismaldo R
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis A Luévano-Martínez
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Monserrat Reyes
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Grecia García-Márquez
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Luis Sobrevia
- Tecnologico de Monterrey, Institute for Obesity Research, School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), São Paulo State University (UNESP), São Paulo, Brazil
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
8
|
Parker J, O’Brien CL, Yeoh C, Gersh FL, Brennecke S. Reducing the Risk of Pre-Eclampsia in Women with Polycystic Ovary Syndrome Using a Combination of Pregnancy Screening, Lifestyle, and Medical Management Strategies. J Clin Med 2024; 13:1774. [PMID: 38541997 PMCID: PMC10971491 DOI: 10.3390/jcm13061774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 05/04/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a multisystem disorder that presents with a variety of phenotypes involving metabolic, endocrine, reproductive, and psychological symptoms and signs. Women with PCOS are at increased risk of pregnancy complications including implantation failure, miscarriage, gestational diabetes, fetal growth restriction, preterm labor, and pre-eclampsia (PE). This may be attributed to the presence of specific susceptibility features associated with PCOS before and during pregnancy, such as chronic systemic inflammation, insulin resistance (IR), and hyperandrogenism, all of which have been associated with an increased risk of pregnancy complications. Many of the features of PCOS are reversible following lifestyle interventions such as diet and exercise, and pregnant women following a healthy lifestyle have been found to have a lower risk of complications, including PE. This narrative synthesis summarizes the evidence investigating the risk of PE and the role of nutritional factors in women with PCOS. The findings suggest that the beneficial aspects of lifestyle management of PCOS, as recommended in the evidence-based international guidelines, extend to improved pregnancy outcomes. Identifying high-risk women with PCOS will allow targeted interventions, early-pregnancy screening, and increased surveillance for PE. Women with PCOS should be included in risk assessment algorithms for PE.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2522, Australia
| | - Claire Louise O’Brien
- Faculty of Science and Technology, University of Canberra, Canberra 2617, Australia;
| | - Christabelle Yeoh
- Next Practice Genbiome, 2/2 New McLean Street, Edgecliff 2027, Australia;
| | - Felice L. Gersh
- College of Medicine, University of Arizona, Tucson, AZ 85004, USA;
| | - Shaun Brennecke
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women’s Hospital, Melbourne 3052, Australia;
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
9
|
Mouzaki M, Woo JG, Divanovic S. Gestational and Developmental Contributors of Pediatric MASLD. Semin Liver Dis 2024; 44:43-53. [PMID: 38423068 DOI: 10.1055/s-0044-1782210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Pediatric metabolic dysfunction-associated steatotic liver disease (MASLD) is common and can be seen as early as in utero. A growing body of literature suggests that gestational and early life exposures modify the risk of MASLD development in children. These include maternal risk factors, such as poor cardiometabolic health (e.g., obesity, gestational diabetes, rapid weight gain during pregnancy, and MASLD), as well as periconceptional dietary exposures, degree of physical activity, intestinal microbiome, and smoking. Paternal factors, such as diet and obesity, also appear to play a role. Beyond gestation, early life dietary exposures, as well as the rate of infant weight gain, may further modify the risk of future MASLD development. The mechanisms linking parental health and environmental exposures to pediatric MASLD are complex and not entirely understood. In conclusion, investigating gestational and developmental contributors to MASLD is critical and may identify future interventional targets for disease prevention.
Collapse
Affiliation(s)
- Marialena Mouzaki
- Divisions of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jessica G Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
10
|
Álvarez D, Ortiz M, Valdebenito G, Crisosto N, Echiburú B, Valenzuela R, Espinosa A, Maliqueo M. Effects of a High-Fat Diet and Docosahexaenoic Acid during Pregnancy on Fatty Acid Composition in the Fetal Livers of Mice. Nutrients 2023; 15:4696. [PMID: 37960348 PMCID: PMC10649644 DOI: 10.3390/nu15214696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
A high-fat diet (HFD) during pregnancy promotes fat accumulation and reduces docosahexaenoic acid (DHA) levels in the liver of the offspring at postnatal ages, which can depend on fetal sex. However, the prenatal mechanisms behind these associations are still unclear. Thus, we analyzed if an HFD alters DHA content and the expression of molecules related to fatty acid (FA) metabolism in the fetal liver. Female C57BL/6 mice were fed a control diet or HFD for 4-6 weeks before pregnancy until the gestational day (GD) 17.5. A subgroup of each diet received DHA (100 mg/Kg) orally from GD 6.5 until 16.5. On GD 17.5, maternal livers, placentas, and livers from male and female fetuses were collected for FA profiling with gas-chromatography and gene expression of molecules related to FA metabolism using qPCR. PPAR-α protein expression was evaluated using Western blot. The gene expression of placental FA transporters was also assessed. An HFD increased eicosapentaenoic acid (EPA) and decreased DHA levels and protein expression of PPAR-α in the fetal livers of both sexes. DHA increased the gene expression of Ppara, Cpt1, and Acsl1 in the livers of female fetuses. Therefore, an HFD reduces DHA levels and PPAR-α, a master regulator of gene expression, in the fetal liver. In turn, the livers of female fetuses seem to be more sensitive to DHA action.
Collapse
Affiliation(s)
- Daniela Álvarez
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
| | - Macarena Ortiz
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
| | - Gabriel Valdebenito
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
- Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Universidad del Desarrollo, Santiago 7610658, Chile
| | - Bárbara Echiburú
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
| | - Rodrigo Valenzuela
- Nutrition Department, School of Medicine, Universidad de Chile, Santiago 8380000, Chile;
| | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile;
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; (D.Á.); (M.O.); (G.V.); (N.C.); (B.E.)
| |
Collapse
|
11
|
Pinto GDA, Murgia A, Lai C, Ferreira CS, Goes VA, Guimarães DDAB, Ranquine LG, Reis DL, Struchiner CJ, Griffin JL, Burton GJ, Torres AG, El-Bacha T. Sphingolipids and acylcarnitines are altered in placentas from women with gestational diabetes mellitus. Br J Nutr 2023; 130:921-932. [PMID: 36539977 DOI: 10.1017/s000711452200397x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gestational diabetes mellitus (GDM) is the most common medical complication of pregnancy and a severe threat to pregnant people and offspring health. The molecular origins of GDM, and in particular the placental responses, are not fully known. The present study aimed to perform a comprehensive characterisation of the lipid species in placentas from pregnancies complicated with GDM using high-resolution MS lipidomics, with a particular focus on sphingolipids and acylcarnitines in a semi-targeted approach. The results indicated that despite no major disruption in lipid metabolism, placentas from GDM pregnancies showed significant alterations in sphingolipids, mostly lower abundance of total ceramides. Additionally, very long-chain ceramides and sphingomyelins with twenty-four carbons were lower, and glucosylceramides with sixteen carbons were higher in placentas from GDM pregnancies. Semi-targeted lipidomics revealed the strong impact of GDM on the placental acylcarnitine profile, particularly lower contents of medium and long-chain fatty-acyl carnitine species. The lower contents of sphingolipids may affect the secretory function of the placenta, and lower contents of long-chain fatty acylcarnitines is suggestive of mitochondrial dysfunction. These alterations in placental lipid metabolism may have consequences for fetal growth and development.
Collapse
Affiliation(s)
- Gabriela D A Pinto
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | | | - Carla Lai
- University of Cagliari, Department of Life and Environmental Science, Cagliari Via Ospedale, Cagliari, Italy
| | - Carolina S Ferreira
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Vanessa A Goes
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Deborah de A B Guimarães
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Layla G Ranquine
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Desirée L Reis
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Claudio J Struchiner
- School of Applied Mathematics, Fundação Getúlio Vargas, Rio de Janeiro, Brazil
- Institute of Social Medicine, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julian L Griffin
- Department of Biochemistry, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alexandre G Torres
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
O'Brien K, Wang Y. The Placenta: A Maternofetal Interface. Annu Rev Nutr 2023; 43:301-325. [PMID: 37603428 DOI: 10.1146/annurev-nutr-061121-085246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The placenta is the gatekeeper between the mother and the fetus. Over the first trimester of pregnancy, the fetus is nourished by uterine gland secretions in a process known as histiotrophic nutrition. During the second trimester of pregnancy, placentation has evolved to the point at which nutrients are delivered to the placenta via maternal blood (hemotrophic nutrition). Over gestation, the placenta must adapt to these variable nutrient supplies, to alterations in maternal physiology and blood flow, and to dynamic changes in fetal growth rates. Numerous questions remain about the mechanisms used to transport nutrients to the fetus and the maternal and fetal determinants of this process. Growing data highlight the ability of the placenta to regulate this process. As new technologies and omics approaches are utilized to study this maternofetal interface, greater insight into this unique organ and its impact on fetal development and long-term health has been obtained.
Collapse
Affiliation(s)
- Kimberly O'Brien
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| | - Yiqin Wang
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| |
Collapse
|
13
|
Easton ZJW, Sarr O, Zhao L, Buzatto AZ, Luo X, Zhao S, Li L, Regnault TRH. An Integrated Multi-OMICS Approach Highlights Elevated Non-Esterified Fatty Acids Impact BeWo Trophoblast Metabolism and Lipid Processing. Metabolites 2023; 13:883. [PMID: 37623828 PMCID: PMC10456680 DOI: 10.3390/metabo13080883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Maternal obesity and gestational diabetes mellitus (GDM) are linked with impaired placental function and early onset of non-communicable cardiometabolic diseases in offspring. Previous studies have highlighted that the dietary non-esterified fatty acids (NEFAs) palmitate (PA) and oleate (OA), key dietary metabolites associated with maternal obesity and GDM, are potential modulators of placental lipid processing. Using the BeWo cell line model, the current study integrated transcriptomic (mRNA microarray), metabolomic, and lipidomic readouts to characterize the underlying impacts of exogenous PA and OA on placental villous trophoblast cell metabolism. Targeted gas chromatography and thin-layer chromatography highlighted that saturated and monounsaturated NEFAs differentially impact BeWo cell lipid profiles. Furthermore, cellular lipid profiles differed when exposed to single and multiple NEFA species. Additional multi-omic analyses suggested that PA exposure is associated with enrichment in β-oxidation pathways, while OA exposure is associated with enrichment in anti-inflammatory and antioxidant pathways. Overall, this study further demonstrated that dietary PA and OA are important regulators of placental lipid metabolism. Encouraging appropriate dietary advice and implementing dietary interventions to maintain appropriate placental function by limiting excessive exposure to saturated NEFAs remain crucial in managing at-risk obese and GDM pregnancies.
Collapse
Affiliation(s)
- Zachary J. W. Easton
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Ousseynou Sarr
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Lin Zhao
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Adriana Zardini Buzatto
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Xian Luo
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Shuang Zhao
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Liang Li
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Timothy R. H. Regnault
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
- Department of Obstetrics and Gynaecology, Western University, B2-401 London Health Science Centre-Victoria Hospital, 800 Commissioners Rd E, London, ON N6H 5W9, Canada
- Children’s Health Research Institute, 800 Commissioners Rd E, London, ON N6C 2V5, Canada
- Lawson Health Research Institute, 750 Base Line Rd E, London, ON N6C 2R5, Canada
| |
Collapse
|
14
|
Diniz MS, Grilo LF, Tocantins C, Falcão-Pires I, Pereira SP. Made in the Womb: Maternal Programming of Offspring Cardiovascular Function by an Obesogenic Womb. Metabolites 2023; 13:845. [PMID: 37512552 PMCID: PMC10386510 DOI: 10.3390/metabo13070845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Obesity incidence has been increasing at an alarming rate, especially in women of reproductive age. It is estimated that 50% of pregnancies occur in overweight or obese women. It has been described that maternal obesity (MO) predisposes the offspring to an increased risk of developing many chronic diseases in an early stage of life, including obesity, type 2 diabetes, and cardiovascular disease (CVD). CVD is the main cause of death worldwide among men and women, and it is manifested in a sex-divergent way. Maternal nutrition and MO during gestation could prompt CVD development in the offspring through adaptations of the offspring's cardiovascular system in the womb, including cardiac epigenetic and persistent metabolic programming of signaling pathways and modulation of mitochondrial metabolic function. Currently, despite diet supplementation, effective therapeutical solutions to prevent the deleterious cardiac offspring function programming by an obesogenic womb are lacking. In this review, we discuss the mechanisms by which an obesogenic intrauterine environment could program the offspring's cardiovascular metabolism in a sex-divergent way, with a special focus on cardiac mitochondrial function, and debate possible strategies to implement during MO pregnancy that could ameliorate, revert, or even prevent deleterious effects of MO on the offspring's cardiovascular system. The impact of maternal physical exercise during an obesogenic pregnancy, nutritional interventions, and supplementation on offspring's cardiac metabolism are discussed, highlighting changes that may be favorable to MO offspring's cardiovascular health, which might result in the attenuation or even prevention of the development of CVD in MO offspring. The objectives of this manuscript are to comprehensively examine the various aspects of MO during pregnancy and explore the underlying mechanisms that contribute to an increased CVD risk in the offspring. We review the current literature on MO and its impact on the offspring's cardiometabolic health. Furthermore, we discuss the potential long-term consequences for the offspring. Understanding the multifaceted effects of MO on the offspring's health is crucial for healthcare providers, researchers, and policymakers to develop effective strategies for prevention and intervention to improve care.
Collapse
Affiliation(s)
- Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
15
|
Yu L, Wang C, Zhang D, Liu M, Liu T, Pan B, Che Q, Liu S, Wang B, Dong X, Guo W. Exosomal circ_0008285 in follicle fluid regulates the lipid metabolism through the miR-4644/ LDLR axis in polycystic ovary syndrome. J Ovarian Res 2023; 16:113. [PMID: 37322492 DOI: 10.1186/s13048-023-01199-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
PURPOSE Exosomal circRNA, as an essential mediator of the follicular microenvironment, has been implicated in the etiological and pathobiological studies of polycystic ovarian syndrome (PCOS). This study aimed to determine abnormal circular RNA (circRNA) expression profiles in follicle fluid (FF) exosomes in patients with PCOS and identify the role of circ_0008285/microRNA (miR)-4644/low-density lipoprotein receptor (LDLR) axis in PCOS. METHODS Sixty-seven women undergoing IVF/ICSI, 31 PCOS patients and 36 non-PCOS patients were included in the cohort study. The circRNA expression profiles of FF exosomes in PCOS (n = 3) and control group (n = 3) were compared by RNA sequencing. In an additional cohort (PCOS:28 vs Control:33), the mRNA expression levels of four circRNAs from FF exosomes were further verified by qRT-PCR. Bioinformatic analysis and dual luciferase reporter gene assay verified the relationship between circ_0008285 and miR-4644 and between miR-4644 and LDLR. KGN cells were infected with sh-circ0008285 and transfected with miR-4644 mimic to verify their roles in lipid metabolism. RESULTS Four circRNAs showed significantly different expressions. Circ_0044234 was overexpressed in PCOS patients, while circ_0006877, circ_0013167 and circ0008285 were decreased in PCOS. Among four differentially expressed circRNAs, circ0008285 was enriched in lipoprotein particle receptor activity and cholesterol metabolism pathway by GO and KEGG pathway analyses. Luciferase assay confirmed the competing endogenous RNA (ceRNA) network circ_0008285/miR-4644 /LDLR. The intercellular experiments on circ_0008285 and its reduction in KGN cells showed that the consumption of circ_0008285 in exosomes could increase the expression of miR-4644 in recipient cells and inhibit the expression of LDLR, as well as increase free fatty acid secretion. CONCLUSION Circ_0008285 can combine with miR-4644 to promote the expression of LDLR and affect the cholesterol metabolism of ovarian granulosa cells in PCOS. Our findings revealed the ceRNA network of circ_0008285 and provided a new path to investigate lipid metabolism abnormalities in PCOS.
Collapse
Affiliation(s)
- Li Yu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China
| | - Chen Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China
| | - Doudou Zhang
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, No. 250 Xiao Mu Qiao Road, 200032, Shanghai, PR China
| | - Miao Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, No. 250 Xiao Mu Qiao Road, 200032, Shanghai, PR China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.725 South Wan Ping Road, 200031, Shanghai, PR China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China
| | - Qi Che
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, No. 250 Xiao Mu Qiao Road, 200032, Shanghai, PR China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, No. 250 Xiao Mu Qiao Road, 200032, Shanghai, PR China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China.
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, No. 250 Xiao Mu Qiao Road, 200032, Shanghai, PR China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China.
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, No. 668 Jin Hu Road, 361015, Xiamen, PR China.
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, No.216 Mudanjiang Road, 200940, Shanghai, PR China.
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China.
- Branch of National Clinical Research Center for Laboratory Medicine, No. 111 Yi Xue Yuan Road, 200032, Shanghai, PR China.
| |
Collapse
|
16
|
Santos ED, Hernández MH, Sérazin V, Vialard F, Dieudonné MN. Human Placental Adaptive Changes in Response to Maternal Obesity: Sex Specificities. Int J Mol Sci 2023; 24:ijms24119770. [PMID: 37298720 DOI: 10.3390/ijms24119770] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Maternal obesity is increasingly prevalent and is associated with elevated morbidity and mortality rates in both mothers and children. At the interface between the mother and the fetus, the placenta mediates the impact of the maternal environment on fetal development. Most of the literature presents data on the effects of maternal obesity on placental functions and does not exclude potentially confounding factors such as metabolic diseases (e.g., gestational diabetes). In this context, the focus of this review mainly lies on the impact of maternal obesity (in the absence of gestational diabetes) on (i) endocrine function, (ii) morphological characteristics, (iii) nutrient exchanges and metabolism, (iv) inflammatory/immune status, (v) oxidative stress, and (vi) transcriptome. Moreover, some of those placental changes in response to maternal obesity could be supported by fetal sex. A better understanding of sex-specific placental responses to maternal obesity seems to be crucial for improving pregnancy outcomes and the health of mothers and children.
Collapse
Affiliation(s)
- Esther Dos Santos
- UFR des Sciences de la Santé Simone Veil, Université de Versailles-Saint Quentin en Yvelines-Université Paris Saclay (UVSQ), INRAE, BREED, F-78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort (EnvA), BREED, F-94700 Maisons-Alfort, France
- Service de Biologie Médicale, Centre Hospitalier de Poissy-Saint Germain, F-78300 Poissy, France
| | - Marta Hita Hernández
- UFR des Sciences de la Santé Simone Veil, Université de Versailles-Saint Quentin en Yvelines-Université Paris Saclay (UVSQ), INRAE, BREED, F-78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort (EnvA), BREED, F-94700 Maisons-Alfort, France
| | - Valérie Sérazin
- UFR des Sciences de la Santé Simone Veil, Université de Versailles-Saint Quentin en Yvelines-Université Paris Saclay (UVSQ), INRAE, BREED, F-78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort (EnvA), BREED, F-94700 Maisons-Alfort, France
- Service de Biologie Médicale, Centre Hospitalier de Poissy-Saint Germain, F-78300 Poissy, France
| | - François Vialard
- UFR des Sciences de la Santé Simone Veil, Université de Versailles-Saint Quentin en Yvelines-Université Paris Saclay (UVSQ), INRAE, BREED, F-78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort (EnvA), BREED, F-94700 Maisons-Alfort, France
- Service de Biologie Médicale, Centre Hospitalier de Poissy-Saint Germain, F-78300 Poissy, France
| | - Marie-Noëlle Dieudonné
- UFR des Sciences de la Santé Simone Veil, Université de Versailles-Saint Quentin en Yvelines-Université Paris Saclay (UVSQ), INRAE, BREED, F-78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort (EnvA), BREED, F-94700 Maisons-Alfort, France
| |
Collapse
|
17
|
Bezerra AF, Alves JPM, Fernandes CCL, Cavalcanti CM, Silva MRL, Conde AJH, Palomino GJQ, Teixeira DÍA, do Rego AC, Rodrigues APR, Rondina D. Impact of high-fat diet consumption during prolonged period of pregnancy on placenta structures and umbilical vascular growth in goats. Anim Reprod 2023; 20:e20230019. [PMID: 37228691 PMCID: PMC10205057 DOI: 10.1590/1984-3143-ar2023-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
This study aimed to verify the impact of high-fat diet consumption for a prolonged period on oxidative stress, fetal growth, umbilical vascular system, and placental structures in pregnant goats. Twenty-two pregnant goats were grouped into the control diet (n= 11) and fat diet (n = 11). Flaxseed meal was added to the fat diet, replacing the corn grain of concentrate, from gestational day 100 to delivery date. Diets were isonitrogenous and isoenergetic, differing in fat content (2.8% vs. 6.3% dry matter). The fat group showed higher feed intake and total plasma lipid levels than the control group (P < 0.001). No difference was found in placentome, and umbilical vascular development. Fat diet-fed goats exhibited a lower systolic peak in the umbilical artery. At delivery, placental traits were similar with the exception of the cotyledon width (P = 0.0075), which was smaller in the fat group and cotyledon surface (P = 0.0047) for multiple pregnancy of fat diet. Cotyledonary epithelium showed more intense staining of lipid droplets and a greater area for lipofuscin staining in the fat group compared to control group (P < 0.001). The mean live weight of the kids was lower in the fat group in the first week after delivery than in control group. Thus, in goats, the continuous administration of a high-fat diet during pregnancy does not appear to modify the fetal-maternal vascular structures but has an impact on a part of the placental structure; therefore, its use must be carefully evaluated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Davide Rondina
- Universidade Estadual do Ceará, Faculdade de Veterinária, Fortaleza, CE, Brasil
| |
Collapse
|
18
|
Yong HEJ, Watkins OC, Mah TKL, Cracknell-Hazra VKB, Pillai RA, Selvam P, Islam MO, Sharma N, Cazenave-Gassiot A, Bendt AK, Wenk MR, Godfrey KM, Lewis RM, Chan SY. Increasing maternal age associates with lower placental CPT1B mRNA expression and acylcarnitines, particularly in overweight women. Front Physiol 2023; 14:1166827. [PMID: 37275238 PMCID: PMC10232777 DOI: 10.3389/fphys.2023.1166827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Older pregnant women have increased risks of complications including gestational diabetes and stillbirth. Carnitine palmitoyl transferase (CPT) expression declines with age in several tissues and is linked with poorer metabolic health. Mitochondrial CPTs catalyze acylcarnitine synthesis, which facilitates fatty acid oxidization as fuel. We hypothesized that the placenta, containing maternally-inherited mitochondria, shows an age-related CPT decline that lowers placental acylcarnitine synthesis, increasing vulnerability to pregnancy complications. We assessed CPT1A, CPT1B, CPT1C and CPT2 mRNA expression by qPCR in 77 placentas and quantified 10 medium and long-chain acylcarnitines by LC-MS/MS in a subset of 50 placentas. Older maternal age associated with lower expression of placental CPT1B, but not CPT1A, CPT1C or CPT2. CPT1B expression positively associated with eight acylcarnitines and CPT1C with three acylcarnitines, CPT1A negatively associated with nine acylcarnitines, while CPT2 did not associate with any acylcarnitine. Older maternal age associated with reductions in five acylcarnitines, only in those with BMI≥ 25 kg/m2, and not after adjusting for CPT1B expression. Our findings suggest that CPT1B is the main transferase for placental long-chain acylcarnitine synthesis, and age-related CPT1B decline may underlie decreased placental metabolic flexibility, potentially contributing to pregnancy complications in older women, particularly if they are overweight.
Collapse
Affiliation(s)
- Hannah E. J. Yong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Oliver C. Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tania K. L. Mah
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Victoria K. B. Cracknell-Hazra
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mohammad O. Islam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anne K. Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R. Wenk
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, United Kingdom
| | - Rohan M. Lewis
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Naomi R, Rusli RNM, Teoh SH, Bahari H, Zakaria ZA. Remodulation Effect of Elateriospermum tapos Yoghurt on Metabolic Profile of Maternal Obesity Induced Cognitive Dysfunction and Anxiety-like Behavior in Female Offspring-An In Vivo Trial on Sprague Dawley Rats. Foods 2023; 12:foods12081613. [PMID: 37107408 PMCID: PMC10137489 DOI: 10.3390/foods12081613] [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: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Pre-pregnancy weight gain induces dysregulation in the metabolic profile of the offspring, thereby serving as a key factor for cognitive decline and anxiety status in the offspring. However, early probiotic supplementation during the gestational period is linked with improved metabolic health. At the same time, a natural plant known as Elateriospermum tapos (E. tapos) is proven to improve cognition and modulate the stress hormone due to its high concentration of flavonoids. However, the effects of medicinal plant integrated probiotics in F1 generations warrants further investigation. Thus, this study aimed to study the effect of E. tapos yoghurt on the maternal obesity induced cognitive dysfunction and anxiety in female offspring. In this study, female Sprague Dawley rats were fed with normal chow (n = 8) or high fat diet (n = 40) across pre-pregnancy, gestation, and weaning. The treatment with different concentrations of E. tapos yoghurt (5, 50, and 500 mg/kg/day) were initiated in the obese dams upon post coitum day 0 up to postnatal day 21 (PND 21). Female offspring were weaned on PND 21 and body mass index, waist circumference, lee index, behavior, metabolic parameter, and antioxidant status were analyzed. The result shows that the female offspring of the 500 mg/kg E. tapos yoghurt supplemented group shows a decreased level of insulin, fasting blood glucose, cholesterol, triglycerides, LDL, low fat tissue mass with a high level of HDL, and an increased level of antioxidant status in the hypothalamus. The behavioral assessment proves that the female offspring of the 500 mg/kg E. tapos yoghurt supplemented group exhibits a high recognition index on novel object/place with low anxiety-like behavior in an open field test. In conclusion, our data signify the beneficial effect of early intervention in obese dams on the transgenerational impact on female offspring's metabolic profile, cognitive performance, and anxiety-like behavior.
Collapse
Affiliation(s)
- Ruth Naomi
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Sabah Universiti Malaysia, Kota Kinabalu 88400, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Rusydatul Nabila Mahmad Rusli
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Sabah Universiti Malaysia, Kota Kinabalu 88400, Malaysia
| |
Collapse
|
20
|
Rasool A, Mahmoud T, O’Tierney-Ginn P. Lipid Aldehydes 4-Hydroxynonenal and 4-Hydroxyhexenal Exposure Differentially Impact Lipogenic Pathways in Human Placenta. BIOLOGY 2023; 12:527. [PMID: 37106728 PMCID: PMC10135722 DOI: 10.3390/biology12040527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Long chain polyunsaturated fatty acids (LCPUFAs), such as the omega-6 (n-6) arachidonic acid (AA) and n-3 docosahexanoic acid (DHA), have a vital role in normal fetal development and placental function. Optimal supply of these LCPUFAs to the fetus is critical for improving birth outcomes and preventing programming of metabolic diseases in later life. Although not explicitly required/recommended, many pregnant women take n-3 LCPUFA supplements. Oxidative stress can cause these LCPUFAs to undergo lipid peroxidation, creating toxic compounds called lipid aldehydes. These by-products can lead to an inflammatory state and negatively impact tissue function, though little is known about their effects on the placenta. Placental exposure to two major lipid aldehydes, 4-hydroxynonenal (4-HNE) and 4-hydroxyhexenal (4-HHE), caused by peroxidation of the AA and DHA, respectively, was examined in the context of lipid metabolism. We assessed the impact of exposure to 25 μM, 50 μM and 100 μM of 4-HNE or 4-HHE on 40 lipid metabolism genes in full-term human placenta. 4-HNE increased gene expression associated with lipogenesis and lipid uptake (ACC, FASN, ACAT1, FATP4), and 4-HHE decreased gene expression associated with lipogenesis and lipid uptake (SREBP1, SREBP2, LDLR, SCD1, MFSD2a). These results demonstrate that these lipid aldehydes differentially affect expression of placental FA metabolism genes in the human placenta and may have implications for the impact of LCPUFA supplementation in environments of oxidative stress.
Collapse
|
21
|
Justesen S, Bilde K, Olesen RH, Pedersen LH, Ernst E, Larsen A. ABCB1 expression is increased in human first trimester placenta from pregnant women classified as overweight or obese. Sci Rep 2023; 13:5175. [PMID: 36997557 PMCID: PMC10063677 DOI: 10.1038/s41598-023-31598-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/14/2023] [Indexed: 04/01/2023] Open
Abstract
Obesity has become a global health challenge also affecting reproductive health. In pregnant women, obesity increases the risk of complications such as preterm birth, macrosomia, gestational diabetes, and preeclampsia. Moreover, obesity is associated with long-term adverse effects for the offspring, including increased risk of cardiovascular and metabolic diseases and neurodevelopmental difficulties. The underlying mechanisms are far from understood, but placental function is essential for pregnancy outcome. Transporter proteins P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) are important for trans-placental transport of endogenous substances like lipids and cortisol, a key hormone in tissue maturation. They also hold a protective function protecting the fetus from xenobiotics (e.g. pharmaceuticals). Animal studies suggest that maternal nutritional status can affect expression of placental transporters, but little is known about the effect on the human placenta, especially in early pregnancy. Here, we investigated if overweight and obesity in pregnant women altered mRNA expression of ABCB1 encoding P-gp or ABCG2 encoding BCRP in first trimester human placenta. With informed consent, 75 first trimester placental samples were obtained from women voluntarily seeking surgical abortion (< gestational week 12) (approval no.: 20060063). Villous samples (average gestational age 9.35 weeks) were used for qPCR analysis. For a subset (n = 38), additional villi were snap-frozen for protein analysis. Maternal BMI was defined at the time of termination of pregnancy. Compared to women with BMI 18.5-24.9 kg/m2 (n = 34), ABCB1 mRNA expression was significantly increased in placenta samples from women classified as overweight (BMI 25-29.9 kg/m2, n = 18) (p = 0.040) and women classified as obese (BMI ≥ 30 kg/m2, n = 23) (p = 0.003). Albeit P-gp expression did not show statistically significant difference between groups, the effect of increasing BMI was the same in male and female pregnancies. To investigate if the P-gp increase was compensated, we determined the expression of ABCG2 which was unaffected by maternal obesity (p = 0.291). Maternal BMI affects ABCB1 but not ABCG2 mRNA expression in first trimester human placenta. Further studies of early placental function are needed to understand how the expression of placental transport proteins is regulated by maternal factors such as nutritional status and determine the potential consequences for placental-fetal interaction.
Collapse
Affiliation(s)
- Signe Justesen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Katrine Bilde
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Rasmus H Olesen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Obstetrics and Gynecology, Randers Regional Hospital, 8930, Randers, Denmark
| | - Lars H Pedersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
- Department of Obstetrics and Gynecology, Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Erik Ernst
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Obstetrics and Gynecology, Horsens Regional Hospital, 8700, Horsens, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| |
Collapse
|
22
|
Ceasrine AM, Devlin BA, Bolton JL, Green LA, Jo YC, Huynh C, Patrick B, Washington K, Sanchez CL, Joo F, Campos-Salazar AB, Lockshin ER, Kuhn C, Murphy SK, Simmons LA, Bilbo SD. Maternal diet disrupts the placenta-brain axis in a sex-specific manner. Nat Metab 2022; 4:1732-1745. [PMID: 36443520 PMCID: PMC10507630 DOI: 10.1038/s42255-022-00693-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
High maternal weight is associated with detrimental outcomes in offspring, including increased susceptibility to neurological disorders such as anxiety, depression and communicative disorders. Despite widespread acknowledgement of sex biases in the development of these disorders, few studies have investigated potential sex-biased mechanisms underlying disorder susceptibility. Here, we show that a maternal high-fat diet causes endotoxin accumulation in fetal tissue, and subsequent perinatal inflammation contributes to sex-specific behavioural outcomes in offspring. In male offspring exposed to a maternal high-fat diet, increased macrophage Toll-like receptor 4 signalling results in excess microglial phagocytosis of serotonin (5-HT) neurons in the developing dorsal raphe nucleus, decreasing 5-HT bioavailability in the fetal and adult brains. Bulk sequencing from a large cohort of matched first-trimester human samples reveals sex-specific transcriptome-wide changes in placental and brain tissue in response to maternal triglyceride accumulation (a proxy for dietary fat content). Further, fetal brain 5-HT levels decrease as placental triglycerides increase in male mice and male human samples. These findings uncover a microglia-dependent mechanism through which maternal diet can impact offspring susceptibility for neuropsychiatric disorder development in a sex-specific manner.
Collapse
Affiliation(s)
- Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Lauren A Green
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Young Chan Jo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Carolyn Huynh
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Bailey Patrick
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Kamryn Washington
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Cristina L Sanchez
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Faith Joo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Elana R Lockshin
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Cynthia Kuhn
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Leigh Ann Simmons
- Department of Human Ecology, Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
23
|
Fernandez-Jimenez N, Fore R, Cilleros-Portet A, Lepeule J, Perron P, Kvist T, Tian FY, Lesseur C, Binder AM, Lozano M, Martorell-Marugán J, Loke YJ, Bakulski KM, Zhu Y, Forhan A, Sammallahti S, Everson TM, Chen J, Michels KB, Belmonte T, Carmona-Sáez P, Halliday J, Daniele Fallin M, LaSalle JM, Tost J, Czamara D, Fernández MF, Gómez-Martín A, Craig JM, Gonzalez-Alzaga B, Schmidt RJ, Dou JF, Muggli E, Lacasaña M, Vrijheid M, Marsit CJ, Karagas MR, Räikkönen K, Bouchard L, Heude B, Santa-Marina L, Bustamante M, Hivert MF, Bilbao JR. A meta-analysis of pre-pregnancy maternal body mass index and placental DNA methylation identifies 27 CpG sites with implications for mother-child health. Commun Biol 2022; 5:1313. [PMID: 36446949 PMCID: PMC9709064 DOI: 10.1038/s42003-022-04267-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Higher maternal pre-pregnancy body mass index (ppBMI) is associated with increased neonatal morbidity, as well as with pregnancy complications and metabolic outcomes in offspring later in life. The placenta is a key organ in fetal development and has been proposed to act as a mediator between the mother and different health outcomes in children. The overall aim of the present work is to investigate the association of ppBMI with epigenome-wide placental DNA methylation (DNAm) in 10 studies from the PACE consortium, amounting to 2631 mother-child pairs. We identify 27 CpG sites at which we observe placental DNAm variations of up to 2.0% per 10 ppBMI-unit. The CpGs that are differentially methylated in placenta do not overlap with CpGs identified in previous studies in cord blood DNAm related to ppBMI. Many of the identified CpGs are located in open sea regions, are often close to obesity-related genes such as GPX1 and LGR4 and altogether, are enriched in cancer and oxidative stress pathways. Our findings suggest that placental DNAm could be one of the mechanisms by which maternal obesity is associated with metabolic health outcomes in newborns and children, although further studies will be needed in order to corroborate these findings.
Collapse
Affiliation(s)
- Nora Fernandez-Jimenez
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU) and Biocruces-Bizkaia Health Research Institute, Leioa, Basque Country Spain
| | - Ruby Fore
- grid.38142.3c000000041936754XDepartment of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
| | - Ariadna Cilleros-Portet
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU) and Biocruces-Bizkaia Health Research Institute, Leioa, Basque Country Spain
| | - Johanna Lepeule
- grid.418110.d0000 0004 0642 0153University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, Grenoble, France
| | - Patrice Perron
- grid.411172.00000 0001 0081 2808Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC Canada
| | - Tuomas Kvist
- grid.7737.40000 0004 0410 2071Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Fu-Ying Tian
- grid.189967.80000 0001 0941 6502Gangarosa Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA USA
| | - Corina Lesseur
- grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alexandra M. Binder
- grid.410445.00000 0001 2188 0957Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI USA ,grid.19006.3e0000 0000 9632 6718Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA USA
| | - Manuel Lozano
- grid.5338.d0000 0001 2173 938XEpidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain ,grid.5338.d0000 0001 2173 938XPreventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Universitat de València, Valencia, Spain
| | - Jordi Martorell-Marugán
- grid.4489.10000000121678994Department of Statistics and Operations Research, University of Granada, Granada, Spain ,grid.4489.10000000121678994Bioinformatics Unit. GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Yuk J. Loke
- grid.1058.c0000 0000 9442 535XMurdoch Children’s Research Institute, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, University of Melbourne, Parkville, VIC Australia
| | - Kelly M. Bakulski
- grid.214458.e0000000086837370Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI USA
| | - Yihui Zhu
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, CA USA
| | - Anne Forhan
- grid.508487.60000 0004 7885 7602Université de Paris, Centre for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Paris, France
| | - Sara Sammallahti
- grid.5645.2000000040459992XDepartment of Child and Adolescent Psychiatry and Psychology, Erasmus MC Rotterdam, The Netherlands
| | - Todd M. Everson
- grid.189967.80000 0001 0941 6502Gangarosa Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Epidemiology, Rollins School of Public health at Emory University, Atlanta, GA USA
| | - Jia Chen
- grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Karin B. Michels
- grid.19006.3e0000 0000 9632 6718Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA USA ,grid.5963.9Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Thalia Belmonte
- grid.411342.10000 0004 1771 1175Health Research Institute of Asturias, ISPA and Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | - Pedro Carmona-Sáez
- grid.4489.10000000121678994Department of Statistics and Operations Research, University of Granada, Granada, Spain ,grid.4489.10000000121678994Bioinformatics Unit. GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Jane Halliday
- grid.1058.c0000 0000 9442 535XMurdoch Children’s Research Institute, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, University of Melbourne, Parkville, VIC Australia
| | - M. Daniele Fallin
- grid.21107.350000 0001 2171 9311Wendy Klag Center for Autism and Developmental Disabilities, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD USA
| | - Janine M. LaSalle
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, CA USA
| | - Jorg Tost
- grid.418135.a0000 0004 0641 3404Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| | - Darina Czamara
- grid.419548.50000 0000 9497 5095Max-Planck-Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Mariana F. Fernández
- grid.4489.10000000121678994University of Granada, Center for Biomedical Research (CIBM), Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.466571.70000 0004 1756 6246CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Antonio Gómez-Martín
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.413740.50000 0001 2186 2871Andalusian School of Public Health (EASP), Granada, Spain
| | - Jeffrey M. Craig
- grid.1058.c0000 0000 9442 535XMurdoch Children’s Research Institute, Parkville, VIC Australia ,grid.1021.20000 0001 0526 7079Deakin University, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Beatriz Gonzalez-Alzaga
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.413740.50000 0001 2186 2871Andalusian School of Public Health (EASP), Granada, Spain
| | - Rebecca J. Schmidt
- grid.27860.3b0000 0004 1936 9684Department of Public Health Sciences and the MIND Institute, University of California Davis School of Medicine, Davis, CA USA
| | - John F. Dou
- grid.214458.e0000000086837370Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI USA
| | - Evelyne Muggli
- grid.1058.c0000 0000 9442 535XMurdoch Children’s Research Institute, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, University of Melbourne, Parkville, VIC Australia
| | - Marina Lacasaña
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.466571.70000 0004 1756 6246CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain ,grid.413740.50000 0001 2186 2871Andalusian School of Public Health (EASP), Granada, Spain
| | - Martine Vrijheid
- grid.466571.70000 0004 1756 6246CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain ,grid.434607.20000 0004 1763 3517ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carmen J. Marsit
- grid.189967.80000 0001 0941 6502Gangarosa Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Epidemiology, Rollins School of Public health at Emory University, Atlanta, GA USA
| | - Margaret R. Karagas
- grid.86715.3d0000 0000 9064 6198Department of Biochemistry and Functional Genomics, Universite de Sherbrooke, Sherbrooke, QC Canada
| | - Katri Räikkönen
- grid.7737.40000 0004 0410 2071Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Luigi Bouchard
- grid.86715.3d0000 0000 9064 6198Department of Biochemistry and Functional Genomics, Universite de Sherbrooke, Sherbrooke, QC Canada ,grid.459278.50000 0004 4910 4652Department of Laboratory Medicine, CIUSSS du Saguenay–Lac-St-Jean – Hôpital Universitaire de Chicoutimi, Chicoutimi, QC Canada
| | - Barbara Heude
- grid.508487.60000 0004 7885 7602Université de Paris, Centre for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Paris, France
| | - Loreto Santa-Marina
- grid.466571.70000 0004 1756 6246CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain ,grid.432380.eBiodonostia, Epidemiology and Public Health Area, Environmental Epidemiology and Child Development Group, 20014 San Sebastian, Basque Country Spain ,Health Department of Basque Government, Sub-directorate of Public Health of Gipuzkoa, San Sebastian, Basque Country Spain
| | - Mariona Bustamante
- grid.466571.70000 0004 1756 6246CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain ,grid.434607.20000 0004 1763 3517ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marie-France Hivert
- grid.38142.3c000000041936754XDepartment of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA ,grid.411172.00000 0001 0081 2808Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC Canada ,grid.32224.350000 0004 0386 9924Diabetes Unit, Massachusetts General Hospital, Boston, MA USA
| | - Jose Ramon Bilbao
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU) and Biocruces-Bizkaia Health Research Institute, Leioa, Basque Country Spain ,grid.512890.7CIBER of diabetes and associated metabolic disorders (CIBERDEM), Madrid, Spain
| |
Collapse
|
24
|
Rasool A, Mahmoud T, Mathyk B, Kaneko-Tarui T, Roncari D, White KO, O’Tierney-Ginn P. Obesity downregulates lipid metabolism genes in first trimester placenta. Sci Rep 2022; 12:19368. [PMID: 36371454 PMCID: PMC9653480 DOI: 10.1038/s41598-022-24040-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
Placentas of obese women have low mitochondrial β-oxidation of fatty acids (FA) and accumulate lipids in late pregnancy. This creates a lipotoxic environment, impairing placental efficiency. We hypothesized that placental FA metabolism is impaired in women with obesity from early pregnancy. We assessed expression of key regulators of FA metabolism in first trimester placentas of lean and obese women. Maternal fasting triglyceride and insulin levels were measured in plasma collected at the time of procedure. Expression of genes associated with FA oxidation (FAO; ACOX1, CPT2, AMPKα), FA uptake (LPL, LIPG, MFSD2A), FA synthesis (ACACA) and storage (PLIN2) were significantly reduced in placentas of obese compared to lean women. This effect was exacerbated in placentas of male fetuses. Placental ACOX1 protein was higher in women with obesity and correlated with maternal circulating triglycerides. The PPARα pathway was enriched for placental genes impacted by obesity, and PPARα antagonism significantly reduced 3H-palmitate oxidation in 1st trimester placental explants. These results demonstrate that obesity and hyperlipidemia impact placental FA metabolism as early as 7 weeks of pregnancy.
Collapse
Affiliation(s)
- Aisha Rasool
- grid.67033.310000 0000 8934 4045Tufts Medical Center, Mother Infant Research Institute, Box# 394, 800 Washington Street, Boston, MA 02111 USA
| | - Taysir Mahmoud
- grid.67033.310000 0000 8934 4045Tufts Medical Center, Mother Infant Research Institute, Box# 394, 800 Washington Street, Boston, MA 02111 USA
| | | | - Tomoko Kaneko-Tarui
- grid.67033.310000 0000 8934 4045Tufts Medical Center, Mother Infant Research Institute, Box# 394, 800 Washington Street, Boston, MA 02111 USA
| | - Danielle Roncari
- grid.67033.310000 0000 8934 4045Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA USA
| | - Katharine O. White
- grid.189504.10000 0004 1936 7558Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston, MA USA
| | - Perrie O’Tierney-Ginn
- grid.67033.310000 0000 8934 4045Tufts Medical Center, Mother Infant Research Institute, Box# 394, 800 Washington Street, Boston, MA 02111 USA ,grid.67033.310000 0000 8934 4045Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA USA ,grid.429997.80000 0004 1936 7531Friedman School of Nutrition, Tufts University, Boston, MA USA
| |
Collapse
|
25
|
Bidne KL, Uhlson C, Palmer C, Berry KZ, Powell TL. Human placental lipid content and lipid metabolic enzyme abundance in obesity and across gestation. Clin Sci (Lond) 2022; 136:1389-1404. [PMID: 36103262 PMCID: PMC10108672 DOI: 10.1042/cs20220479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Changes in placental lipid metabolism influence the delivery of lipids critical for fetal development and fetal requirements for lipids change across gestation. We hypothesized that placental lipid content and metabolic enzyme protein levels increase across gestation and are elevated in obesity. Placentas (4-40 weeks' gestation) were collected from control (body mass index, BMI = 18.5-24.9, n=37) and obese (BMI > 30, n=19) pregnant women. Trophoblast villous tissue was homogenized and subjected to liquid chromatography tandem mass spectrometry (LC-MS/MS) for phospholipid and triacylglycerol (TAG) analysis and western blot for protein quantification. The placental content of TAG species and nine of 35 identified phosphatidylcholines (PC) were significantly higher (P<0.05) in first trimester (28-79%, 10-47%, respectively). Furthermore, two TAG and three PC differed by maternal BMI and were significantly increased (P<0.05) in the obese group in first trimester (72-87%, 88-119%, respectively). Placental protein abundance of glycerol-2-phosphate (GPAT3) and 1-acyl-sn-glycerol-3-phosphate acyltransferase 2 (AGPAT2), involved in de novo synthesis of PC and TAG, were higher (P<0.05) in the first trimester (66 and 74%, respectively). The protein abundance of the PC-remodeling enzyme PLA2G4c was also higher (63%) in first trimester (P<0.05). In conclusion, the placental content of many phospholipid and TAG species and the protein level of associated synthesis enzymes are higher in first-trimester human placenta. The high PC content may be related to the rapid membrane expansion in early pregnancy and the low placental oxygen tension may promote the accumulation of tissue TAGs in first trimester. Maternal obesity had only limited impact on placental lipid content and metabolic enzyme protein abundance.
Collapse
Affiliation(s)
- Katie L. Bidne
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charis Uhlson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire Palmer
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karin Zemski Berry
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
26
|
Anam AK, Cooke KM, Dratver MB, O'Bryan JV, Perley LE, Guller SM, Hwang JJ, Taylor HS, Goedeke L, Kliman HJ, Vatner DF, Flannery CA. Insulin increases placental triglyceride as a potential mechanism for fetal adiposity in maternal obesity. Mol Metab 2022; 64:101574. [PMID: 35970449 PMCID: PMC9440306 DOI: 10.1016/j.molmet.2022.101574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Maternal obesity increases the incidence of excess adiposity in newborns, resulting in lifelong diabetes risk. Elevated intrauterine fetal adiposity has been attributed to maternal hyperglycemia; however, this hypothesis does not account for the increased adiposity seen in newborns of mothers with obesity who have euglycemia. We aimed to explore the placental response to maternal hyperinsulinemia and the effect of insulin-like growth factor 2 (IGF-2) in promoting fetal adiposity by increasing storage and availability of nutrients to the fetus. METHODS We used placental villous explants and isolated trophoblasts from normal weight and obese women to assess the effect of insulin and IGF-2 on triglyceride content and insulin receptor signaling. Stable isotope tracer methods were used ex vivo to determine effect of hormone treatment on de novo lipogenesis (DNL), fatty acid uptake, fatty acid oxidation, and esterification in the placenta. RESULTS Here we show that placentae from euglycemic women with normal weight and obesity both have abundant insulin receptor. Placental depth and triglyceride were greater in women with obesity compared with normal weight women. In syncytialized placental trophoblasts and villous explants, insulin and IGF-2 activate insulin receptor, induce expression of lipogenic transcription factor SREBP-1 (sterol regulatory element-binding protein 1), and stimulate triglyceride accumulation. We demonstrate elevated triglyceride is attributable to increased esterification of fatty acids, without contribution from DNL and without an acceleration of fatty acid uptake. CONCLUSIONS Our work reveals that obesity-driven aberrations in maternal metabolism, such as hyperinsulinemia, alter placental metabolism in euglycemic conditions, and may explain the higher prevalence of excess adiposity in the newborns of obese women.
Collapse
Affiliation(s)
- Anika K Anam
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Katherine M Cooke
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Milana Bochkur Dratver
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Jane V O'Bryan
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Lauren E Perley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Seth M Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Janice J Hwang
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Leigh Goedeke
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Harvey J Kliman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Daniel F Vatner
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Clare A Flannery
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Brombach C, Tong W, Giussani DA. Maternal obesity: new placental paradigms unfolded. Trends Mol Med 2022; 28:823-835. [PMID: 35760668 DOI: 10.1016/j.molmed.2022.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 01/24/2023]
Abstract
The prevalence of maternal obesity is increasing at an alarming rate, and is providing a major challenge for obstetric practice. Adverse effects on maternal and fetal health are mediated by complex interactions between metabolic, inflammatory, and oxidative stress signaling in the placenta. Endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) are common downstream pathways of cell stress, and there is evidence that this conserved homeostatic response may be a key mediator in the pathogenesis of placental dysfunction. We summarize the current literature on the placental cellular and molecular changes that occur in obese women. A special focus is cast onto placental ER stress in obese pregnancy, which may provide a novel link for future investigation.
Collapse
Affiliation(s)
| | - Wen Tong
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge CB2 3EL, Cambridge UK.
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge CB2 3EL, Cambridge UK; Cambridge Cardiovascular Centre for Research Excellence, Cambridge CB2 0QQ, UK.
| |
Collapse
|
28
|
Rojas-Rodriguez R, Price LL, Somogie J, Hauguel-de Mouzon S, Kalhan SC, Catalano PM. Maternal Lipid Metabolism Is Associated With Neonatal Adiposity: A Longitudinal Study. J Clin Endocrinol Metab 2022; 107:e3759-e3768. [PMID: 35686573 PMCID: PMC9387706 DOI: 10.1210/clinem/dgac360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Pregnancy is characterized by progressive decreases in glucose insulin sensitivity. Low insulin sensitivity resulting in hyperglycemia is associated with higher neonatal adiposity. However, less is known regarding lipid metabolism, particularly lipid insulin sensitivity in pregnancy and neonatal adiposity. OBJECTIVE Because higher maternal prepregnancy body mass index is strongly associated with both hyperlipidemia and neonatal adiposity, we aimed to examine the longitudinal changes in basal and clamp maternal lipid metabolism as contributors to neonatal adiposity. METHODS Twelve women planning a pregnancy were evaluated before pregnancy, in early (12-14 weeks), and late (34-36 weeks) gestation. Body composition was estimated using hydrodensitometry. Basal and hyperinsulinemic-euglycemic clamp glucose and glycerol turnover (GLYTO) were measured using 2H2-glucose and 2H5-glycerol and substrate oxidative/nonoxidative metabolism with indirect calorimetry. Total body electrical conductivity was used to estimate neonatal body composition. RESULTS Basal free-fatty acids decreased with advancing gestation (P = 0.0210); however, basal GLYTO and nonoxidative lipid metabolism increased over time (P = 0.0046 and P = 0.0052, respectively). Further, clamp GLYTO and lipid oxidation increased longitudinally over time (P = 0.0004 and P = 0.0238, respectively). There was a median 50% increase and significant positive correlation during both basal and clamp GLYTO from prepregnancy through late gestation. Neonatal adiposity correlated with late pregnancy basal and clamp GLYTO (r = 0.6515, P = 0.0217; and r = 0.6051, P = 0.0371). CONCLUSIONS Maternal prepregnancy and late pregnancy measures of basal and clamp lipid metabolism are highly correlated. Late pregnancy basal and clamp GLYTO are significantly associated with neonatal adiposity and account for ~40% of the variance in neonatal adiposity. These data emphasize the importance of maternal lipid metabolism relating to fetal fat accrual.
Collapse
Affiliation(s)
| | - Lori Lyn Price
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
- Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA 02111, USA
| | - Jessica Somogie
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | | | - Satish C Kalhan
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Patrick M Catalano
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
29
|
Wang M, Wang Z, Miao Y, Wei H, Peng J, Zhou Y. Diallyl Trisulfide Promotes Placental Angiogenesis by Regulating Lipid Metabolism and Alleviating Inflammatory Responses in Obese Pregnant Mice. Nutrients 2022; 14:nu14112230. [PMID: 35684030 PMCID: PMC9182607 DOI: 10.3390/nu14112230] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
The placental tissue serves as an exchanger between the mother and the fetus during pregnancy in mammals. Proper placental angiogenesis is central to the health of both the mother and the growth and development of the fetus. Maternal obesity is associated with impaired placental function, resulting in restricted placental blood vessel development and fetal developmental disorders. Hydrogen sulfide (H2S) is a ubiquitous second messenger in cells that has many biological effects such as promoting angiogenesis, anti-inflammation, anti-oxidation and promoting lipid metabolism. However, in the case of maternal obesity, whether H2S can be used as an important signaling molecule to regulate body metabolism, alleviate placental inflammation levels and promote placental angiogenesis is still unclear. In this study, diallyl trisulfide (DATS), which is a well-known H2S donor, was derived from garlic and used to treat obese pregnant mice induced by a high-fat diet, to determine its effects on lipid metabolism and inflammation, as well as placental morphology and placental angiogenesis. Here, we show that DATS treatment increased litter size and alive litter size. DATS improved the H2S level in the serum and placenta of the mice. In addition, DATS treatment improved insulin resistance and lipid metabolism, reduced the inflammatory response and alleviated placental vascular dysplasia caused by obesity in obese mice. In summary, our research revealed that H2S is an important signaling molecule in vivo, which can regulate placental angiogenesis and improve the reproductive performance in maternal obesity. The addition of H2S donor DATS during pregnancy promoted placental angiogenesis by regulating lipid metabolism and alleviating inflammatory responses in obese pregnant mice.
Collapse
Affiliation(s)
- Miaomiao Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
| | - Zhaoyu Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
| | - Yueyue Miao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Correspondence: (J.P.); (Y.Z.)
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.W.); (Z.W.); (Y.M.); (H.W.)
- Correspondence: (J.P.); (Y.Z.)
| |
Collapse
|
30
|
Zhang L, Wang Z, Wu H, Gao Y, Zheng J, Zhang J. Maternal High-Fat Diet Impairs Placental Fatty Acid β-Oxidation and Metabolic Homeostasis in the Offspring. Front Nutr 2022; 9:849684. [PMID: 35495939 PMCID: PMC9050107 DOI: 10.3389/fnut.2022.849684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 12/30/2022] Open
Abstract
Maternal overnutrition can affect fetal growth and development, thus increasing susceptibility to obesity and diabetes in later life of the offspring. Placenta is the central organ connecting the developing fetus with the maternal environment. It is indicated placental fatty acid metabolism plays an essential role in affecting the outcome of the pregnancy and fetus. However, the role of placental fatty acid β-oxidation (FAO) in maternal overnutrition affecting glucose metabolism in the offspring remains unclear. In this study, C57BL/6J female mice were fed with normal chow or high-fat diet before and during pregnancy and lactation. The placenta and fetal liver were collected at gestation day 18.5, and the offspring's liver was collected at weaning. FAO-related genes and AMP-activated protein kinase (AMPK) signaling pathway were examined both in the placenta and in the human JEG-3 trophoblast cells. FAO-related genes were further examined in the liver of the fetuses and in the offspring at weaning. We found that dams fed with high-fat diet showed higher fasting blood glucose, impaired glucose tolerance at gestation day 14.5 and higher serum total cholesterol (T-CHO) at gestation day 18.5. The placental weight and lipid deposition were significantly increased in maternal high-fat diet group. At weaning, the offspring mice of high-fat diet group exhibited higher body weight, impaired glucose tolerance, insulin resistance and increased serum T-CHO, compared with control group. We further found that maternal high-fat diet downregulated mRNA and protein expressions of carnitine palmitoyltransferase 2 (CPT2), a key enzyme in FAO, by suppressing the AMPK/Sirt1/PGC1α signaling pathway in the placenta. In JEG-3 cells, protein expressions of CPT2 and CPT1b were both downregulated by suppressing the AMPK/Sirt1/PGC1α signaling pathway under glucolipotoxic condition, but were later restored by the AMPK agonist 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR). However, there was no difference in CPT2 and CPT1 gene expression in the liver of fetuses and offspring at weaning age. In conclusion, maternal high-fat diet can impair gene expression involved in FAO in the placenta by downregulating the AMPK signaling pathway, and can cause glucose and lipid dysfunction of offspring at weaning, indicating that placental FAO may play a crucial role in regulating maternal overnutrition and metabolic health in the offspring.
Collapse
|
31
|
Hufnagel A, Dearden L, Fernandez-Twinn DS, Ozanne SE. Programming of cardiometabolic health: the role of maternal and fetal hyperinsulinaemia. J Endocrinol 2022; 253:R47-R63. [PMID: 35258482 PMCID: PMC9066586 DOI: 10.1530/joe-21-0332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022]
Abstract
Obesity and gestational diabetes during pregnancy have multiple short- and long-term consequences for both mother and child. One common feature of pregnancies complicated by maternal obesity and gestational diabetes is maternal hyperinsulinaemia, which has effects on the mother and her adaptation to pregnancy. Even though insulin does not cross the placenta insulin can act on the placenta as well affecting placental growth, angiogenesis and lipid metabolism. Obese and gestational diabetic pregnancies are often characterised by maternal hyperglycaemia resulting in exposure of the fetus to high levels of glucose, which freely crosses the placenta. This leads to stimulation of fetal ß-cells and insulin secretion in the fetus. Fetal hyperglycaemia/hyperinsulinaemia has been shown to cause multiple complications in fetal development, such as altered growth trajectories, impaired neuronal and cardiac development and early exhaustion of the pancreas. These changes could increase the susceptibility of the offspring to develop cardiometabolic diseases later in life. In this review, we aim to summarize and review the mechanisms by which maternal and fetal hyperinsulinaemia impact on (i) maternal health during pregnancy; (ii) placental and fetal development; (iii) offspring energy homeostasis and long-term cardiometabolic health; (iv) how interventions can alleviate these effects.
Collapse
Affiliation(s)
- Antonia Hufnagel
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Laura Dearden
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| |
Collapse
|
32
|
Napso T, Lean SC, Lu M, Mort EJ, Desforges M, Moghimi A, Bartels B, El‐Bacha T, Fowden AL, Camm EJ, Sferruzzi‐Perri AN. Diet-induced maternal obesity impacts feto-placental growth and induces sex-specific alterations in placental morphology, mitochondrial bioenergetics, dynamics, lipid metabolism and oxidative stress in mice. Acta Physiol (Oxf) 2022; 234:e13795. [PMID: 35114078 PMCID: PMC9286839 DOI: 10.1111/apha.13795] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023]
Abstract
AIM The current study investigated the impact of maternal obesity on placental phenotype in relation to fetal growth and sex. METHODS Female C57BL6/J mice were fed either a diet high in fat and sugar or a standard chow diet, for 6 weeks prior to, and during, pregnancy. At day 19 of gestation, placental morphology and mitochondrial respiration and dynamics were assessed using high-resolution respirometry, stereology, and molecular analyses. RESULTS Diet-induced maternal obesity increased the rate of small for gestational age fetuses in both sexes, and increased blood glucose concentrations in offspring. Placental weight, surface area, and maternal blood spaces were decreased in both sexes, with reductions in placental trophoblast volume, oxygen diffusing capacity, and an increased barrier to transfer in males only. Despite these morphological changes, placental mitochondrial respiration was unaffected by maternal obesity, although the influence of fetal sex on placental respiratory capacity varied between dietary groups. Moreover, in males, but not females, maternal obesity increased mitochondrial complexes (II and ATP synthase) and fission protein DRP1 abundance. It also reduced phosphorylated AMPK and capacity for lipid synthesis, while increasing indices of oxidative stress, specifically in males. In females only, placental mitochondrial biogenesis and capacity for lipid synthesis, were both enhanced. The abundance of uncoupling protein-2 was decreased by maternal obesity in both fetal sexes. CONCLUSION Maternal obesity exerts sex-dependent changes in placental phenotype in association with alterations in fetal growth and substrate supply. These findings may inform the design of personalized lifestyle interventions or therapies for obese pregnant women.
Collapse
Affiliation(s)
- Tina Napso
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Samantha C. Lean
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Minhui Lu
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Mort
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Michelle Desforges
- Division of Developmental Biology and Medicine Maternal & Fetal Health Research Centre University of Manchester Manchester UK
| | - Ali Moghimi
- The Children’s Hospital at Westmead Westmead New South Wales Australia
- Department of Paediatrics Monash University Monash Victoria Australia
| | - Beverly Bartels
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Tatiana El‐Bacha
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Abigail L. Fowden
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Camm
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Amanda N. Sferruzzi‐Perri
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| |
Collapse
|
33
|
Song L, Wang N, Peng Y, Sun B, Cui W. Placental lipid transport and content in response to maternal overweight and gestational diabetes mellitus in human term placenta. Nutr Metab Cardiovasc Dis 2022; 32:692-702. [PMID: 35109996 DOI: 10.1016/j.numecd.2021.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Placental lipid transport is altered in women with high prepregnancy body mass index (pre-BMI) or gestational diabetes (GDM), which consequently affects foetal growth. However, the interaction of maternal overweight (OW) and GDM on placental lipid metabolism and possible adaptations are less studied. We aimed to examine whether maternal OW or GDM is the main factor disrupting placental lipid processing in human term placenta. METHODS AND RESULTS A total of 152 lean (18.5 ≤ pre-BMI ≤ 23.9 kg/m2) and OW (24 ≤ pre-BMI ≤ 27.9 kg/m2) pregnant women with or without GDM with a scheduled delivery by caesarean section were recruited. Maternal venous blood samples were used to measure metabolic parameters during pregnancy. Term placentas and cord blood were collected at delivery to determine placental lipid metabolism and foetal circulating lipid levels. Maternal OW significantly increased the placental mRNA expression of genes involved in lipid metabolism (FAT/CD36, FATP1, FATP4, FATP6, and PPAR-α), elevated placental lipid content (triglyceride, cholesterol), enhanced placental mTORC1-rpS6 and ERK1/2 signalling, increased cord blood insulin levels and birth weight. Neonatal birth weight was positively correlated with maternal pre-BMI, placental ERK1/2 signalling and cord blood insulin. There was an interaction between OW and GDM in regulating key placental fuel transport and storage gene expression (LPL, FATP6, FABP7, PPAR-α, PPAR-β, PPAR-γ, IR-β, GLUT1, SNAT2, SNAT4, and LAT1). CONCLUSION Maternal OW mainly affects placental lipid metabolism, which may contribute to foetal overgrowth and may impact long-term offspring health. GDM plays a less significant role in affecting placental lipid transfer and other mechanisms may be involved.
Collapse
Affiliation(s)
- Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ning Wang
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanqi Peng
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Wei Cui
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
34
|
Bukowski MR, Singh BB, Roemmich JN, Claycombe-Larson KJ. Lipidomic Analysis of TRPC1 Ca 2+-Permeable Channel-Knock Out Mouse Demonstrates a Vital Role in Placental Tissue Sphingolipid and Triacylglycerol Homeostasis Under Maternal High-Fat Diet. Front Endocrinol (Lausanne) 2022; 13:854269. [PMID: 35360063 PMCID: PMC8960927 DOI: 10.3389/fendo.2022.854269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The transient receptor potential canonical channel 1 (TRPC1) is a ubiquitous Ca2+-permeable integral membrane protein present in most tissues, including adipose and placenta, and functionally regulates energetic homeostasis. We demonstrated that elimination of TRPC1 in a mouse model increased body adiposity and limited adipose accumulation under a high fat diet (HFD) even under conditions of exercise. Additionally, intracellular Ca2+ regulates membrane lipid content via the activation of the protein kinase C pathway, which may impact placental membrane lipid content and structure. Based upon this we investigated the effect of HFD and TRPC1 elimination on neutral lipids (triacylglycerol and cholesteryl ester), membrane lipids (phosphatidylcholine and phosphatidylethanolamine), and other multifunctional lipid species (unesterified cholesterol, sphingomyelins, ceramides). The concentration of unesterified cholesterol and sphingomyelin increased with gestational age (E12.5 to E 18.5.) indicating possible increases in plasma membrane fluidity. Diet-dependent increases ceramide concentration at E12.5 suggest a pro-inflammatory role for HFD in early gestation. TRPC1-dependent decreases in cholesterol ester concentration with concomitant increases in long-chain polyunsaturated fatty acid -containing triacylglycerols indicate a disruption of neutral lipid homeostasis that may be tied to Ca2+ regulation. These results align with changes in lipid content observed in studies of preeclamptic human placenta.
Collapse
Affiliation(s)
- Michael R. Bukowski
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND, United States
- *Correspondence: Michael R. Bukowski, ; Kate J. Claycombe-Larson,
| | - Brij B. Singh
- School of Dentistry, UT Health Science Center San Antonio, San Antonio, TX, United States
| | - James N. Roemmich
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND, United States
| | - Kate J. Claycombe-Larson
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND, United States
- *Correspondence: Michael R. Bukowski, ; Kate J. Claycombe-Larson,
| |
Collapse
|
35
|
Song Y, Lyu C, Li M, Rahman ML, Chen Z, Zhu Y, Hinkle SN, Chen L, Mitro SD, Li LJ, Weir NL, Tsai MY, Zhang C. Plasma Acylcarnitines during Pregnancy and Neonatal Anthropometry: A Longitudinal Study in a Multiracial Cohort. Metabolites 2021; 11:metabo11120885. [PMID: 34940643 PMCID: PMC8704426 DOI: 10.3390/metabo11120885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
As surrogate readouts reflecting mitochondrial dysfunction, elevated levels of plasma acylcarnitines have been associated with cardiometabolic disorders, such as obesity, gestational diabetes, and type 2 diabetes. This study aimed to examine prospective associations of acylcarnitine profiles across gestation with neonatal anthropometry, including birthweight, birthweight z score, body length, sum of skinfolds, and sum of body circumferences. We quantified 28 acylcarnitines using electrospray ionization tandem mass spectrometry in plasma collected at gestational weeks 10–14, 15–26, 23–31, and 33–39 among 321 pregnant women from the National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies-Singletons. A latent-class trajectory approach was applied to identify trajectories of acylcarnitines across gestation. We examined the associations of individual acylcarnitines and distinct trajectory groups with neonatal anthropometry using weighted generalized linear models adjusting for maternal age, race/ethnicity, education, parity, gestational age at blood collection, and pre-pregnancy body mass index (BMI). We identified three distinct trajectory groups in C2, C3, and C4 and two trajectory groups in C5, C10, C5–DC, C8:1, C10:1, and C12, respectively. Women with nonlinear decreasing C12 levels across gestation (5.7%) had offspring with significantly lower birthweight (−475 g; 95% CI, −942, −6.79), birthweight z score (−0.39, −0.71, −0.06), and birth length (−1.38 cm, −2.49, −0.27) than those with persistently stable C12 levels (94.3%) (all nominal p value < 0.05). Women with consistently higher levels of C10 (6.1%) had offspring with thicker sum of skinfolds (4.91 mm, 0.85, 8.98) than did women with lower levels (93.9%) during pregnancy, whereas women with lower C10:1 levels (12.6%) had offspring with thicker sum of skinfolds (3.23 mm, 0.19, 6.27) than did women with abruptly increasing levels (87.4%) (p < 0.05). In conclusion, this study suggests that distinctive trajectories of C10, C10:1, and C12 acylcarnitine levels throughout pregnancy were significantly associated with neonatal anthropometry.
Collapse
Affiliation(s)
- Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN 46202, USA;
| | - Chen Lyu
- Department of Population Health, Division of Biostatistics, NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA;
| | - Ming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
| | - Mohammad L. Rahman
- Department of Population Medicine and Harvard Pilgrim Healthcare Institute, Harvard Medical School, Boston, MA 02215, USA;
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD 20817, USA; (Z.C.); (S.D.M.)
| | - Yeyi Zhu
- Kaiser Permanente Northern California Division of Research, Oakland, CA 94612, USA;
| | - Stefanie N. Hinkle
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Liwei Chen
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA;
| | - Susanna D. Mitro
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD 20817, USA; (Z.C.); (S.D.M.)
| | - Ling-Jun Li
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Natalie L. Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Cuilin Zhang
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD 20817, USA; (Z.C.); (S.D.M.)
- Correspondence: ; Tel.: +1-301-435-6917
| |
Collapse
|
36
|
Tarui T, Rasool A, O'Tierney-Ginn P. How the placenta-brain lipid axis impacts the nutritional origin of child neurodevelopmental disorders: Focus on attention deficit hyperactivity disorder and autism spectrum disorder. Exp Neurol 2021; 347:113910. [PMID: 34742689 DOI: 10.1016/j.expneurol.2021.113910] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/19/2021] [Accepted: 10/31/2021] [Indexed: 12/01/2022]
Abstract
Dietary fish is a rich source of omega-3 (n-3) fatty acids, and as such, is believed to have played an important role in the evolution of the human brain and its advanced cognitive function. The long chain polyunsaturated fatty acids, particularly the n-3 docosahexanoic acid (DHA), are critical for proper neurological development and function. Both low plasma DHA and obesity in pregnancy are associated with neurodevelopmental disorders such as attention deficit and hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) in childhood, and n-3 supplementation has been shown to improve symptoms, as reviewed herein. The mechanisms underlying the connection between maternal obesity, n-3 fatty acid levels and offspring's neurological outcomes are poorly understood, but we review the evidence for a mediating role of the placenta in this relationship. Despite promising data that n-3 fatty acid supplementation mitigates the effect of maternal obesity on placental lipid metabolism, few clinical trials or animal studies have considered the neurological outcomes of offspring of mothers with obesity supplemented with n-3 FA in pregnancy.
Collapse
Affiliation(s)
- Tomo Tarui
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States of America
| | - Aisha Rasool
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States of America
| | - Perrie O'Tierney-Ginn
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States of America.
| |
Collapse
|
37
|
Peng J, Yang M, Li G, Zhang X, Huang Y, Tang Y. Effects of palmitic acid and eicosapentaenoic acid on angiogenesis of porcine vascular endothelial cells. Vet Med Sci 2021; 7:2260-2267. [PMID: 34547178 PMCID: PMC8604142 DOI: 10.1002/vms3.577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Restricted placental angiogenesis is an important cause of intrauterine growth retardation in piglets. During pregnancy, sow obesity can result in an increase in placental lipid deposition, subsequently inhibiting placental angiogenesis and fetal development. However, the effect of different types of fatty acids on placental angiogenesis is still unclear. Trophoblast cells and vascular endothelial cells constitute two important types of placental tissue. In this study, we used palmitic acid (C16:0) and eicosapentaenoic acid (C20:5, n-3), respectively, to treat porcine trophectoderm cells (pTr2) and porcine iliac artery endothelial cells (PIEC) to study the effects of saturated fatty acids and n-3 polyunsaturated fatty acids (PUFAs) on placental angiogenesis in vitro. We found that C16:0 caused significant cytotoxicity in pTr2 and PIEC (p < 0.01) and inhibited the proliferation and migration of PIEC (p < 0.01), whereas C20:5 treatment exhibited very low cytotoxicity and minimal inhibition of cellular proliferation. Meanwhile, a low concentration of C16:0 had no effect on the tube formation in PIEC, whereas C20:5 significantly promoted tube formation of PIEC (p < 0.01). These results suggested that saturated fatty acids and n-3 PUFAs had different effects on placental angiogenesis. As essential functional fatty acid, n-3 PUFA might be effective measure in alleviating the placental lipotoxicity caused by sow obesity during pregnancy.
Collapse
Affiliation(s)
- Jie Peng
- Innovative Institute of Animal Healthy BreedingZhongkai University of Agriculture and EngineeringGuangzhouChina
- College of Animal Sciences and TechnologyZhongkai University of Agriculture and EngineeringGuangzhouChina
| | - Menglin Yang
- Innovative Institute of Animal Healthy BreedingZhongkai University of Agriculture and EngineeringGuangzhouChina
- College of Animal Sciences and TechnologyZhongkai University of Agriculture and EngineeringGuangzhouChina
| | - Guoli Li
- Guangzhou Fishtech Biotechnology Co., Ltd.GuangzhouChina
| | - Xiu Zhang
- Ministry of Agriculture and Rural AffairsWENS Research InstituteYunfuChina
| | - Yanhua Huang
- Innovative Institute of Animal Healthy BreedingZhongkai University of Agriculture and EngineeringGuangzhouChina
- College of Animal Sciences and TechnologyZhongkai University of Agriculture and EngineeringGuangzhouChina
- Guangzhou Fishtech Biotechnology Co., Ltd.GuangzhouChina
| | - Yimei Tang
- Innovative Institute of Animal Healthy BreedingZhongkai University of Agriculture and EngineeringGuangzhouChina
- College of Light Industry and Food SciencesZhongkai University of Agriculture and EngineeringGuangzhouChina
| |
Collapse
|
38
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
39
|
Uhl O, Lewis RM, Hirschmugl B, Crozier S, Inskip H, Gazquez A, Harvey NC, Cooper C, Desoye G, Koletzko B, Wadsack C, Demmelmair H, Godfrey KM. Placental polar lipid composition is associated with placental gene expression and neonatal body composition. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158971. [PMID: 34029703 DOI: 10.1016/j.bbalip.2021.158971] [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: 10/13/2020] [Revised: 05/05/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022]
Abstract
The polar-lipid composition of the placenta reflects its cellular heterogeneity and metabolism. This study explored relationships between placental polar-lipid composition, gene expression and neonatal body composition. Placental tissue and maternal and offspring data were collected in the Southampton Women's Survey. Lipid and RNA were extracted from placental tissue and polar lipids measured by mass spectrometry, while gene expression was assessed using the nCounter analysis platform. Principal component analysis was used to identify patterns within placental lipid composition and these were correlated with neonatal body composition and placental gene expression. In the analysis of placental lipids, the first three principal components explained 19.1%, 12.7% and 8.0% of variation in placental lipid composition, respectively. Principal component 2 was characterised by high principal component scores for acyl-alkyl-glycerophosphatidylcholines and lipid species containing DHA. Principal component 2 was associated with placental weight and neonatal lean mass; this component was associated with gene expression of APOE, PLIN2, FATP2, FABP4, LEP, G0S2, PNPLA2 and SRB1. Principal components 1 and 3 were not related to birth outcomes but they were associated with the gene expression of lipid related genes. Principal component 1 was associated with expression of LEP, APOE, FATP2 and ACAT2. Principal component 3 was associated with expression of PLIN2, PLIN3 and PNPLA2. This study demonstrates that placentas of different sizes have specific differences in polar-lipid composition and related gene expression. These differences in lipid composition were associated with birth weight and neonatal lean mass, suggesting that placental lipid composition may influence prenatal lean mass accretion.
Collapse
Affiliation(s)
- Olaf Uhl
- Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospitals, Ludwig-Maximilians-Universität Munich, Germany
| | | | - Birgit Hirschmugl
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Sarah Crozier
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; NIHR Applied Research Collaboration Wessex, Southampton Science Park, UK.
| | - Hazel Inskip
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Antonio Gazquez
- Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospitals, Ludwig-Maximilians-Universität Munich, Germany; University of Murcia, Department of Physiology, Murcia, Spain.
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.
| | - Berthold Koletzko
- Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospitals, Ludwig-Maximilians-Universität Munich, Germany.
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Hans Demmelmair
- Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospitals, Ludwig-Maximilians-Universität Munich, Germany.
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
40
|
Belcastro L, Ferreira CS, Saraiva MA, Mucci DB, Murgia A, Lai C, Vigor C, Oger C, Galano JM, Pinto GDA, Griffin JL, Torres AG, Durand T, Burton GJ, Sardinha FLC, El-Bacha T. Decreased Fatty Acid Transporter FABP1 and Increased Isoprostanes and Neuroprostanes in the Human Term Placenta: Implications for Inflammation and Birth Weight in Maternal Pre-Gestational Obesity. Nutrients 2021; 13:2768. [PMID: 34444927 PMCID: PMC8398812 DOI: 10.3390/nu13082768] [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: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/26/2022] Open
Abstract
The rise in prevalence of obesity in women of reproductive age in developed and developing countries might propagate intergenerational cycles of detrimental effects on metabolic health. Placental lipid metabolism is disrupted by maternal obesity, which possibly affects the life-long health of the offspring. Here, we investigated placental lipid metabolism in women with pre-gestational obesity as a sole pregnancy complication and compared it to placental responses of lean women. Open profile and targeted lipidomics were used to assess placental lipids and oxidised products of docosahexaenoic (DHA) and arachidonic acid (AA), respectively, neuroprostanes and isoprostanes. Despite no overall signs of lipid accumulation, DHA and AA levels in placentas from obese women were, respectively, 2.2 and 2.5 times higher than those from lean women. Additionally, a 2-fold increase in DHA-derived neuroprostanes and a 1.7-fold increase in AA-derived isoprostanes were seen in the obese group. These changes correlated with a 70% decrease in placental FABP1 protein. Multivariate analyses suggested that neuroprostanes and isoprostanes are associated with maternal and placental inflammation and with birth weight. These results might shed light on the molecular mechanisms associated with altered placental fatty acid metabolism in maternal pre-gestational obesity, placing these oxidised fatty acids as novel mediators of placental function.
Collapse
Affiliation(s)
- Livia Belcastro
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.B.); (M.A.S.); (D.B.M.)
| | - Carolina S. Ferreira
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (C.S.F.); (G.D.A.P.); (A.G.T.)
| | - Marcelle A. Saraiva
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.B.); (M.A.S.); (D.B.M.)
| | - Daniela B. Mucci
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.B.); (M.A.S.); (D.B.M.)
| | - Antonio Murgia
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK; (A.M.); (J.L.G.)
| | - Carla Lai
- Department of Environmental and Life Sciences, University of Cagliari, 09124 Cagliari, Italy;
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 Route de Mende, 34293 Montpellier, France; (C.V.); (C.O.); (J.-M.G.); (T.D.)
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 Route de Mende, 34293 Montpellier, France; (C.V.); (C.O.); (J.-M.G.); (T.D.)
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 Route de Mende, 34293 Montpellier, France; (C.V.); (C.O.); (J.-M.G.); (T.D.)
| | - Gabriela D. A. Pinto
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (C.S.F.); (G.D.A.P.); (A.G.T.)
| | - Julian L. Griffin
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK; (A.M.); (J.L.G.)
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2BX, UK
| | - Alexandre G. Torres
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (C.S.F.); (G.D.A.P.); (A.G.T.)
- Lipid Biochemistry and Lipidomics Laboratory, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 Route de Mende, 34293 Montpellier, France; (C.V.); (C.O.); (J.-M.G.); (T.D.)
| | - Graham J. Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK;
| | - Fátima L. C. Sardinha
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.B.); (M.A.S.); (D.B.M.)
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (C.S.F.); (G.D.A.P.); (A.G.T.)
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK;
| |
Collapse
|
41
|
Watkins OC, Selvam P, Appukuttan Pillai R, Cracknell-Hazra VKB, Yong HEJ, Sharma N, Cazenave-Gassiot A, Bendt AK, Godfrey KM, Lewis RM, Wenk MR, Chan SY. Placental 13C-DHA metabolism and relationship with maternal BMI, glycemia and birthweight. Mol Med 2021; 27:84. [PMID: 34362294 PMCID: PMC8349043 DOI: 10.1186/s10020-021-00344-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fetal docosahexaenoic acid (DHA) supply relies on preferential transplacental transfer, which is regulated by placental DHA lipid metabolism. Maternal hyperglycemia and obesity associate with higher birthweight and fetal DHA insufficiency but the role of placental DHA metabolism is unclear. METHODS Explants from 17 term placenta were incubated with 13C-labeled DHA for 48 h, at 5 or 10 mmol/L glucose treatment, and the production of 17 individual newly synthesized 13C-DHA labeled lipids quantified by liquid chromatography mass spectrometry. RESULTS Maternal BMI positively associated with 13C-DHA-labeled diacylglycerols, triacylglycerols, lysophospholipids, phosphatidylcholine and phosphatidylethanolamine plasmalogens, while maternal fasting glycemia positively associated with five 13C-DHA triacylglycerols. In turn, 13C-DHA-labeled phospholipids and triacylglycerols positively associated with birthweight centile. In-vitro glucose treatment increased most 13C-DHA-lipids, but decreased 13C-DHA phosphatidylethanolamine plasmalogens. However, with increasing maternal BMI, the magnitude of the glucose treatment induced increase in 13C-DHA phosphatidylcholine and 13C-DHA lysophospholipids was curtailed, with further decline in 13C-DHA phosphatidylethanolamine plasmalogens. Conversely, with increasing birthweight centile glucose treatment induced increases in 13C-DHA triacylglycerols were exaggerated, while glucose treatment induced decreases in 13C-DHA phosphatidylethanolamine plasmalogens were diminished. CONCLUSIONS Maternal BMI and glycemia increased the production of different placental DHA lipids implying impact on different metabolic pathways. Glucose-induced elevation in placental DHA metabolism is moderated with higher maternal BMI. In turn, findings of associations between many DHA lipids with birthweight suggest that BMI and glycemia promote fetal growth partly through changes in placental DHA metabolism.
Collapse
Affiliation(s)
- Oliver C Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Victoria K B Cracknell-Hazra
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anne K Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Rohan M Lewis
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
42
|
Rocha-Gomes A, Teixeira AE, de Oliveira DG, Santiago CMO, da Silva AA, Riul TR, Lacerda ACR, Mendonça VA, Rocha-Vieira E, Leite HR. LPS tolerance prevents anxiety-like behavior and amygdala inflammation of high-fat-fed dams' adolescent offspring. Behav Brain Res 2021; 411:113371. [PMID: 34019914 DOI: 10.1016/j.bbr.2021.113371] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/15/2021] [Accepted: 05/15/2021] [Indexed: 02/04/2023]
Abstract
Maternal high-fat diets (HFD) can generate inflammation in the offspring's amygdala, which can lead to anxiety-like behaviors. Conversely, lipopolysaccharide (LPS) tolerance can reduce neuroinflammation in the offspring caused by maternal high-fat diets. This study evaluated the combination of LPS tolerance and high-fat maternal diet on amygdala's inflammatory parameters and the anxiety-like behavior in adolescent offspring. Female pregnant Wistar rats received randomly a standard diet or a high-fat diet during gestation and lactation. On gestation days 8, 10, and 12, half of the females in each group were intraperitonially injected with LPS (0.1 mg.kg-1). After weaning, the male offspring (n = 96) were placed in individual boxes in standard conditions, and when 6 weeks-old, the animals underwent: Open-Field, Light/Dark Box, Elevated Plus-Maze, and Rotarod tests. When 50 days-old the offspring were euthanized and the amygdala removed for cytokine and redox status analysis. The offspring in the HFD group showed lower amygdala IL-10 levels, high IL-6/IL-10 ratio, and anxiety-like behaviors. These effects were attenuated in the HFD offspring submitted to LPS tolerance, which showed an anti-inflammatory compensatory response in the amygdala. Also, this group showed a higher activity of the enzyme catalase in the amygdala. In addition, receiving the combination of LPS tolerance and maternal HFD did not lead to anxiety-like behavior in the offspring. The results suggest that LPS tolerance attenuated amygdala inflammation through an anti-inflammatory compensatory response besides preventing anxiety-like behavior caused by the high-fat maternal diet.
Collapse
Affiliation(s)
- Arthur Rocha-Gomes
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Dalila Gomes de Oliveira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Ana Cristina Rodrigues Lacerda
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Vanessa Amaral Mendonça
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Etel Rocha-Vieira
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Faculdade de Medicina do Campus JK, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Hércules Ribeiro Leite
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós Graduação em Ciências da Reabilitação, Universidade Federal de Minas Gerais, Diamantina, MG, Brazil.
| |
Collapse
|
43
|
Overweight and obesity in pregnancy: their impact on epigenetics. Eur J Clin Nutr 2021; 75:1710-1722. [PMID: 34230629 PMCID: PMC8636269 DOI: 10.1038/s41430-021-00905-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/12/2021] [Accepted: 03/16/2021] [Indexed: 02/02/2023]
Abstract
Over the last few decades, the prevalence of obesity has risen to epidemic proportions worldwide. Consequently, the number of obesity in pregnancy has risen drastically. Gestational overweight and obesity are associated with impaired outcomes for mother and child. Furthermore, studies show that maternal obesity can lead to long-term consequences in the offspring, increasing the risk for obesity and cardiometabolic disease in later life. In addition to genetic mechanisms, mounting evidence demonstrates the induction of epigenetic alterations by maternal obesity, which can affect the offspring’s phenotype, thereby influencing the later risk of obesity and cardiometabolic disease. Clear evidence in this regard comes from various animal models of maternal obesity. Evidence derived from clinical studies remains limited. The current article gives an overview of pathophysiological changes associated with maternal obesity and their consequences on placental structure and function. Furthermore, a short excurse is given on epigenetic mechanisms and emerging data regarding a putative interaction between metabolism and epigenetics. Finally, a summary of important findings of animal and clinical studies investigating maternal obesity-related epigenetic effects is presented also addressing current limitations of clinical studies.
Collapse
|
44
|
Continuous glucose monitoring in obese pregnant women with no hyperglycemia on glucose tolerance test. PLoS One 2021; 16:e0253047. [PMID: 34111215 PMCID: PMC8191902 DOI: 10.1371/journal.pone.0253047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/27/2021] [Indexed: 11/27/2022] Open
Abstract
Objective The objective of the present study was to compare 24-hour glycemic levels between obese pregnant women with normal glucose tolerance and non-obese pregnant women. Methods In the present observational, longitudinal study, continuous glucose monitoring was performed in obese pregnant women with normal oral glucose tolerance test with 75 g of glucose between the 24th and the 28th gestational weeks. The control group (CG) consisted of pregnant women with normal weight who were selected by matching the maternal age and parity with the same characteristics of the obese group (OG). Glucose measurements were obtained during 72 hours. Results Both the groups were balanced in terms of baseline characteristics (age: 33.5 [28.7–36.0] vs. 32.0 [26.0–34.5] years, p = 0.5 and length of pregnancy: 25.0 [24.0–25.0] vs. 25.5 [24.0–28.0] weeks, p = 0.6 in the CG and in the OG, respectively). Pre-breakfast glycemic levels were 77.77 ± 10.55 mg/dL in the CG and 82.02 ± 11.06 mg/dL in the OG (p<0.01). Glycemic levels at 2 hours after breakfast were 87.31 ± 13.10 mg/dL in the CG and 93.48 ± 18.74 mg/dL in the OG (p<0.001). Daytime blood glucose levels were 87.6 ± 15.4 vs. 93.1 ± 18.3 mg/dL (p<0.001) and nighttime blood glucose levels were 79.3 ± 15.8 vs. 84.7 ± 16.3 mg/dL (p<0.001) in the CG and in the OG, respectively. The 24-hour, daytime, and nighttime values of the area under the curve were higher in the OG when compared with the CG (85.1 ± 0.16 vs. 87.9 ± 0.12, 65.6 ± 0.14 vs. 67.5 ± 0.10, 19.5 ± 0.07 vs. 20.4 ± 0.05, respectively; p<0.001). Conclusion The results of the present study showed that obesity in pregnancy was associated with higher glycemic levels even in the presence of normal findings on glucose tolerance test.
Collapse
|
45
|
Fowden AL, Camm EJ, Sferruzzi-Perri AN. Effects of Maternal Obesity On Placental Phenotype. Curr Vasc Pharmacol 2021; 19:113-131. [PMID: 32400334 DOI: 10.2174/1570161118666200513115316] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/26/2022]
Abstract
The incidence of obesity is rising rapidly worldwide with the consequence that more women are entering pregnancy overweight or obese. This leads to an increased incidence of clinical complications during pregnancy and of poor obstetric outcomes. The offspring of obese pregnancies are often macrosomic at birth although there is also a subset of the progeny that are growth-restricted at term. Maternal obesity during pregnancy is also associated with cardiovascular, metabolic and endocrine dysfunction in the offspring later in life. As the interface between the mother and fetus, the placenta has a central role in programming intrauterine development and is known to adapt its phenotype in response to environmental conditions such as maternal undernutrition and hypoxia. However, less is known about placental function in the abnormal metabolic and endocrine environment associated with maternal obesity during pregnancy. This review discusses the placental consequences of maternal obesity induced either naturally or experimentally by increasing maternal nutritional intake and/or changing the dietary composition. It takes a comparative, multi-species approach and focusses on placental size, morphology, nutrient transport, metabolism and endocrine function during the later stages of obese pregnancy. It also examines the interventions that have been made during pregnancy in an attempt to alleviate the more adverse impacts of maternal obesity on placental phenotype. The review highlights the potential role of adaptations in placental phenotype as a contributory factor to the pregnancy complications and changes in fetal growth and development that are associated with maternal obesity.
Collapse
Affiliation(s)
- A L Fowden
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom
| | - E J Camm
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom
| | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom
| |
Collapse
|
46
|
Barrett E, Loverin A, Wang H, Carlson M, Larsen TD, Almeida MM, Whitman J, Baack ML, Joss-Moore LA. Uteroplacental Insufficiency with Hypoxia Upregulates Placental PPARγ-KMT5A Axis in the Rat. Reprod Sci 2021; 28:1476-1488. [PMID: 33398850 PMCID: PMC8215892 DOI: 10.1007/s43032-020-00434-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/13/2020] [Indexed: 12/30/2022]
Abstract
The placenta represents a critical node in fetal lipid acquisition, yet the mechanisms by which the placenta handles lipids under normal and pathologic conditions are incompletely understood. A key player in placental lipid handling is peroxisome proliferator-activated receptor gamma (PPARγ). PPARγ influences global gene expression via its regulation of the epigenetic modifier lysine methyltransferase 5A (KMT5A), which places a methyl group on histone 4 lysine 20 (H4K20me) of target genes. Here we test the hypothesis that KMT5A is present in both the human and rat placentas and is affected by uteroplacental insufficiency (UPI) in the rat in association with increased placental lipid accumulation. We assessed levels and localization of KMT5A, as well as lipid droplet accumulation, in human placental tissue collected from maternal donors after delivery by planned cesarean section. Using a rat model of UPI, we also evaluated the effects of UPI on lipid accumulation, PPARγ, KMT5A, and H4K20me in the rat placenta. In this study, we show for the first time the presence and activity of KMT5A, in human and in rat placentas. We also demonstrate that in the rat placenta, UPI increases hypoxia, KMT5a expression, and activity in association with increased lipid accumulation in placenta supporting male fetuses. Placental PPARγ-KMT5A axis may be an important mediator of placental lipid handling.
Collapse
Affiliation(s)
- Emily Barrett
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, 84109, USA
| | - Amy Loverin
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, 84109, USA
| | - Haimei Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA
| | | | - Tricia D Larsen
- Environmental Influences on Health and Disease, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Mariana M Almeida
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jenna Whitman
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA
| | - Michelle L Baack
- Environmental Influences on Health and Disease, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Lisa A Joss-Moore
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA.
| |
Collapse
|
47
|
Natarajan SK, Bruett T, Muthuraj PG, Sahoo PK, Power J, Mott JL, Hanson C, Anderson-Berry A. Saturated free fatty acids induce placental trophoblast lipoapoptosis. PLoS One 2021; 16:e0249907. [PMID: 33886600 PMCID: PMC8062006 DOI: 10.1371/journal.pone.0249907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Obesity during pregnancy increases the risk for maternal complications like gestational diabetes, preeclampsia, and maternal inflammation. Maternal obesity also increases the risk of childhood obesity, intrauterine growth restriction (IUGR) and diabetes to the offspring. Increased circulating free fatty acids (FFAs) in obesity due to adipose tissue lipolysis induces lipoapoptosis to hepatocytes, cholangiocytes, and pancreatic-β-cells. During the third trimester of human pregnancy, there is an increase in maternal lipolysis and release of FFAs into the circulation. It is currently unknown if increased FFAs during gestation as a result of maternal obesity cause placental cell lipoapoptosis. Increased exposure of FFAs during maternal obesity has been shown to result in placental lipotoxicity. The objective of the present study is to determine saturated FFA-induced trophoblast lipoapoptosis and also to test the protective role of monounsaturated fatty acids against FFA-induced trophoblast lipoapoptosis using in vitro cell culture model. Here, we hypothesize that saturated FFAs induce placental trophoblast lipoapoptosis, which was prevented by monounsaturated fatty acids. METHODS Biochemical and structural markers of apoptosis by characteristic nuclear morphological changes with DAPI staining, and caspase 3/7 activity was assessed. Cleaved PARP and cleaved caspase 3 were examined by western blot analysis. RESULTS Treatment of trophoblast cell lines, JEG-3 and JAR cells with palmitate (PA) or stearate (SA) induces trophoblast lipoapoptosis as evidenced by a significant increase in apoptotic nuclear morphological changes and caspase 3/7 activity. We observed that saturated FFAs caused a concentration-dependent increase in placental trophoblast lipoapoptosis. We also observed that monounsaturated fatty acids like palmitoleate and oleate mitigates placental trophoblast lipoapoptosis caused due to PA exposure. CONCLUSION We show that saturated FFAs induce trophoblast lipoapoptosis. Co-treatment of monounsaturated fatty acids like palmitoleate and oleate protects against FFA-induced trophoblast lipoapoptosis.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail:
| | - Taylor Bruett
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Prakash K. Sahoo
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Jillian Power
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Corrine Hanson
- College of Allied Health Professions Medical Nutrition Education, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
48
|
Nogues P, Dos Santos E, Couturier-Tarrade A, Berveiller P, Arnould L, Lamy E, Grassin-Delyle S, Vialard F, Dieudonne MN. Maternal Obesity Influences Placental Nutrient Transport, Inflammatory Status, and Morphology in Human Term Placenta. J Clin Endocrinol Metab 2021; 106:e1880-e1896. [PMID: 32936881 DOI: 10.1210/clinem/dgaa660] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023]
Abstract
CONTEXT Maternal obesity has a significant impact on placental development. However, this impact on the placenta's structure and function (ie, nutrient transport and hormone and cytokine production) is a controversial subject. OBJECTIVE We hypothesized that maternal obesity is associated with morphologic, secretory, and nutrient-related changes and elevated levels of inflammation in the placenta. DESIGN We collected samples of placental tissue from 2 well-defined groups of pregnant women from 2017 to 2019. We compared the 2 groups regarding placental cytokine and hormone secretion, immune cell content, morphology, and placental nutrient transporter expressions. SETTING Placenta were collected after caesarean section performed by experienced clinicians at Centre Hospitalier Intercommunal (CHI) of Poissy-Saint-Germain-en-Laye. PATIENTS The main inclusion criteria were an age between 27 and 37 years old, no complications of pregnancy, and a first-trimester body mass index of 18-25 kg/m2 for the nonobese (control) group and 30-40 kg/m2 for the obese group. RESULTS In contrast to our starting hypothesis, we observed that maternal obesity was associated with (1) lower placental IL-6 expression and macrophage/leukocyte infiltration, (2) lower placental expression of GLUT1 and SNAT1-2, (3) a lower placental vessel density, and (4) lower levels of placental leptin and human chorionic gonadotropin production. CONCLUSION These results suggest that the placenta is a plastic organ and could optimize fetal growth. A better understanding of placental adaptation is required because these changes may partly determine the fetal outcome in cases of maternal obesity.
Collapse
Affiliation(s)
- Perrine Nogues
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Esther Dos Santos
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
- Service de Biologie Médicale, Centre Hospitalier Intercommunal de Poissy-Saint-Germain, Poissy, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Paul Berveiller
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
- Service de Gynécologie-Obstétrique, Centre Hospitalier Intercommunal de Poissy-Saint-Germain-en-Laye, Poissy, France
| | - Lucie Arnould
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Elodie Lamy
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, Département de Biotechnologie de la Santé, Montigny le Bretonneux, France
| | - Stanislas Grassin-Delyle
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, Département de Biotechnologie de la Santé, Montigny le Bretonneux, France
- Hôpital Foch, Département des maladies des voies respiratoires, Suresnes, France
| | - François Vialard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
- Département de Biologie de la Reproduction, Cytogénétique, Gynécologie et Obstétrique, Centre Hospitalier Intercommunal de Poissy-Saint-Germain-en-Laye, Poissy, France
| | - Marie-Noëlle Dieudonne
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| |
Collapse
|
49
|
Huang K, Deng J, Yang Y, Qiao N, Zhang Z, Li Q, Han Q, Zhang H, Yang F, Ji Y, Cui J, Ali F, Li Y, Tang Z. Effects of 25(OH)D 3 supplementation during late gestation on the serum biochemistry and reproductive performance of aged sows and newborn piglets. J Anim Physiol Anim Nutr (Berl) 2021; 105:908-915. [PMID: 33713505 DOI: 10.1111/jpn.13530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 12/20/2020] [Accepted: 02/15/2021] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to investigate the effects of diet type (normal or low Ca and P diets) and 25(OH)D3 supplementation (with or with not 2000 IU/kg 25(OH)D3 ) during late gestation on the serum biochemistry and reproductive performance of aged sows and newborn piglets. A total of 40 sows, which are at their 7th parity, were divided into four groups: control group (standard diet), low Ca group, 25(OH)D3 group and low Ca plus 25(OH)D3 group respectively (10 in each group). The blood of sows on day 100 and 114 of gestation and newborn piglets was collected for serum biochemical analyses. Results showed that the reproductive performance of sows was not influenced by diet type or 25(OH)D3 supplementation (p > 0.05). And the addition of 25(OH)D3 to diet low Ca group caused that the content of serum TG in sows on day 100 of gestation was not different from that of the control group (p > 0.05). The addition of 25(OH)D3 significantly decreases the content of serum TG in sows on day 114 of gestation (p < 0.05). The addition of 25(OH)D3 significantly increased the content of serum UREA and CREA in newborn piglets (p < 0.05). Overall, feeding 2000 IU/kg 25(OH)D3 to aged sows at late gestation had no effects on reproductive performance, but partly contributed to keeping serum TG balance in sows and may indicate increased pressure on kidneys in newborn piglets.
Collapse
Affiliation(s)
- Kunxuan Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jichang Deng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yanyang Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Na Qiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhuowei Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Quanwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fan Yang
- Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yanju Ji
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jianxin Cui
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Farah Ali
- University College of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
50
|
Sundrani DP, Karkhanis AR, Joshi SR. Peroxisome Proliferator-Activated Receptors (PPAR), fatty acids and microRNAs: Implications in women delivering low birth weight babies. Syst Biol Reprod Med 2021; 67:24-41. [PMID: 33719831 DOI: 10.1080/19396368.2020.1858994] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Low birth weight (LBW) babies are associated with neonatal morbidity and mortality and are at increased risk for noncommunicable diseases (NCDs) in later life. However, the molecular determinants of LBW are not well understood. Placental insufficiency/dysfunction is the most frequent etiology for fetal growth restriction resulting in LBW and placental epigenetic processes are suggested to be important regulators of pregnancy outcome. Early life exposures like altered maternal nutrition may have long-lasting effects on the health of the offspring via epigenetic mechanisms like DNA methylation and microRNA (miRNA) regulation. miRNAs have been recognized as major regulators of gene expression and are known to play an important role in placental development. Angiogenesis in the placenta is known to be regulated by transcription factor peroxisome proliferator-activated receptor (PPAR) which is activated by ligands such as long-chain-polyunsaturated fatty acids (LCPUFA). In vitro studies in different cell types indicate that fatty acids can influence epigenetic mechanisms like miRNA regulation. We hypothesize that maternal fatty acid status may influence the miRNA regulation of PPAR genes in the placenta in women delivering LBW babies. This review provides an overview of miRNAs and their regulation of PPAR gene in the placenta of women delivering LBW babies.Abbreviations: AA - Arachidonic Acid; Ago2 - Argonaute2; ALA - Alpha-Linolenic Acid; ANGPTL4 - Angiopoietin-Like Protein 4; C14MC - Chromosome 14 miRNA Cluster; C19MC - Chromosome 19 miRNA Cluster; CLA - Conjugated Linoleic Acid; CSE - Cystathionine γ-Lyase; DHA - Docosahexaenoic Acid; EFA - Essential Fatty Acids; E2F3 - E2F transcription factor 3; EPA - Eicosapentaenoic Acid; FGFR1 - Fibroblast Growth Factor Receptor 1; GDM - Gestational Diabetes Mellitus; hADMSCs - Human Adipose Tissue-Derived Mesenchymal Stem Cells; hBMSCs - Human Bone Marrow Mesenchymal Stem Cells; HBV - Hepatitis B Virus; HCC - Hepatocellular Carcinoma; HCPT - Hydroxycamptothecin; HFD - High-Fat Diet; Hmads - Human Multipotent Adipose-Derived Stem; HSCS - Human Hepatic Stellate Cells; IUGR - Intrauterine Growth Restriction; LA - Linoleic Acid; LBW - Low Birth Weight; LCPUFA - Long-Chain Polyunsaturated Fatty Acids; MEK1 - Mitogen-Activated Protein Kinase 1; MiRNA - MicroRNA; mTOR - Mammalian Target of Rapamycin; NCDs - NonCommunicable Diseases; OA - Oleic Acid; PASMC - Pulmonary Artery Smooth Muscle Cell; PLAG1 - Pleiomorphic Adenoma Gene 1; PPAR - Peroxisome Proliferator-Activated Receptor; PPARα - PPAR alpha; PPARγ - PPAR gamma; PPARδ - PPAR delta; pre-miRNA - precursor miRNA; RISC - RNA-Induced Silencing Complex; ROS - Reactive Oxygen Species; SAT - Subcutaneous Adipose Tissue; WHO - World Health Organization.
Collapse
Affiliation(s)
- Deepali P Sundrani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Aishwarya R Karkhanis
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|