1
|
Ye J, Shi R, Wu X, Fan H, Zhao Y, Hu X, Wang L, Bo X, Li D, Ge Y, Wang D, Xia B, Zhao Z, Xiao C, Zhao B, Wang Y, Liu X. Stevioside mitigates metabolic dysregulation in offspring induced by maternal high-fat diet: the role of gut microbiota-driven thermogenesis. Gut Microbes 2025; 17:2452241. [PMID: 39838262 DOI: 10.1080/19490976.2025.2452241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/08/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Maternal obesity poses a significant threat to the metabolic profiles of offspring. Microorganisms acquired from the mother early in life critically affect the host's metabolic functions. Natural non-nutritive sweeteners, particularly stevioside (STV), play a crucial role in reducing obesity and affecting gut microbiota composition. Based on this, we hypothesized that maternal STV supplementation could improve the health of mothers and offspring by altering their gut microbiota. Our study found that maternal STV supplementation reduced obesity during pregnancy, decreased abnormal lipid accumulation in offspring mice caused by maternal obesity, and modified the gut microbiota of both dams and offspring, notably increasing the abundance of Lactobacillus apodemi (L. apodemi). Co-housing and fecal microbiota transplant experiments confirmed that gut microbiota mediated the effects of STV on metabolic disorders. Furthermore, treatment with L. apodemi alone replicated the beneficial effects of STV, which were associated with increased thermogenesis. In summary, maternal STV supplementation could alleviate lipid metabolic disorders in offspring by enhancing L. apodemi levels and promoting thermogenic activity, potentially involving changes in bile acid metabolism pathways.
Collapse
Affiliation(s)
- Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
- Institute of Biology, Gansu Academy of Sciences, Lanzhou, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Xiaoning Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Hua Fan
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yapei Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xinyun Hu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Lulu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaowei Bo
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Dongning Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yunshu Ge
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Bing Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhenting Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Chunxia Xiao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yutang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
2
|
Peterson AK, Zhu Y, Feng J, Calafat AM, Hedderson MM, Alexeeff SE, Ferrara A. Urinary concentrations of early and mid-pregnancy parabens and gestational diabetes: A nested case-control study within the PETALS cohort. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 974:179253. [PMID: 40154079 DOI: 10.1016/j.scitotenv.2025.179253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Parabens are widely used preservatives with endocrine-disrupting properties, but their role in glucose metabolism during pregnancy is unclear. This study examines prospective associations between urinary concentrations of four parabens in early and mid-pregnancy and gestational diabetes (GDM). A matched case-control study nested within a diverse longitudinal pregnancy cohort (PETALS) with universal GDM screening matched GDM cases to two controls (111 cases; 222 controls). Urine samples collected 2015-2017 in early (14 ± 2.3 weeks) and mid-pregnancy (20 ± 2.4 weeks) were analyzed for paraben concentrations with mass spectrometry. Area-under-the-time-concentration-curve (AUC) assessed cumulative exposure. Conditional logistic regression models evaluated associations between paraben concentrations and GDM, adjusting for covariates. We a priori examined effect modification by Asian/Pacific Islander (A/PI) race/ethnicity due to the case-control matching and GDM prevalence highest among A/PI. Participants were 31 ± 5 years old and 40 % A/PI, 33 % Hispanic, 14 % White and 9 % Black. Methylparaben and propylparaben had >94 % detection, while ethylparaben and butylparaben ranged from 22 %-51 %. Paraben exposure was not associated with GDM overall. Among A/PI, higher methylparaben concentrations exhibited higher odds of GDM: early-pregnancy OR 1.14 per IQR (95 % CI: 0.89,1.45) and AUC 1.07 (0.89,1.30) compared to non-A/PI (early-pregnancy 0.81 [0.62,1.06] and AUC 0.70 [0.44,1.12]; Pinteraction = 0.01 and 0.03, respectively). A/PI mid-pregnancy ethylparaben exposure (detectable vs non-detectable) was linked to higher GDM odds (2.00 [0.84,4.76] vs. non-A/PI 0.47 [0.17,1.27]; Pinteraction = 0.04) as was mid-pregnancy propylparaben exposure (Tertile 2 vs. 1: 3.67 [1.21,11.1] vs. non-A/PI 0.70 [0.22, 2.25]; Pinteraction = 0.04). Although overall paraben exposure was not associated with GDM, interactions by A/PI race/ethnicity suggested potential increased odds of GDM related to propylparaben, methylparaben, and ethylparaben exposure. Future studies should explore paraben exposure in diverse populations.
Collapse
Affiliation(s)
- Alicia K Peterson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America; Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America.
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America; Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America
| | - Juanran Feng
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Monique M Hedderson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America; Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America
| | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America
| | - Assiamira Ferrara
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America; Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, United States of America
| |
Collapse
|
3
|
Phengpol N, Promsan S, Pengrattanachot N, Jaruan O, Sutthasupha P, Lungkaphin A. Maternal obesity promotes impaired renal autophagic process and kidney injury in male offspring. Int J Obes (Lond) 2025:10.1038/s41366-025-01751-3. [PMID: 40133698 DOI: 10.1038/s41366-025-01751-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Obesity during pregnancy increases the risk of obesity, insulin resistance, diabetes, and the development and progression of chronic kidney disease (CKD) in later life in offspring. Impaired renal autophagic process is linked to kidney dysfunction in the setting of increased renal lipid accumulation. The aim of this study was to elucidate the effect of maternal obesity on kidney injury related to impaired renal autophagic process in the offspring. METHODS Maternal obesity model was conducted using female C57BL/6 mice fed with high-fat diet (HFD) for 8 weeks before mating. HFD was consecutively maintained throughout gestation and lactation. Male offspring were selected for investigation after weaning. Metabolic parameters and kidney morphology were performed. Renal insulin signaling, lipid metabolism, lipid accumulation, fibrosis and autophagy were determined. RESULTS Male offspring of HFD fed mothers developed obesity with insulin resistance, hyperglycemia, hyperlipidemia and consequently promoted kidney injury. Maternal obesity increased CD36, FAS, SREBP1c and Perilipin-2 while suppressed PPARα and CPT1A. The reduction of AMPK, SIRT1, Beclin-1, LC3B, and LAMP2 and the elevation of mTOR and SQSTM1/P62 were observed. These findings indicated the impairment of autophagy and renal lipid metabolism exaggerating renal lipid accumulation in the offspring of maternal obesity. CONCLUSIONS This study demonstrated that long-term HFD consumption in mothers promoted obesity with insulin resistance related kidney injury through the impairment of autophagic process and renal lipid metabolism in the offspring. These circumstances accelerated kidney injury and contributed to an increased susceptibility to CKD in male offspring of maternal obesity.
Collapse
Affiliation(s)
- Nichakorn Phengpol
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasivimon Promsan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Onanong Jaruan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prempree Sutthasupha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Functional Foods for Health and Disease, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Functional Food Research Center for Well-being, Multidisciplinary Research Institute Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
4
|
Heeren FAN, Himadi M, Flood-Grady E, Xu K, Loop MS, Francois M, Louis-Jacques AF, Thompson L, Cardel MI, Gillespie Y, DeCicco L, Lemas DJ. Recruitment and retention of participants with obesity into a longitudinal birth cohort: the Breastfeeding and Early Child Health (BEACH) study. Int J Obes (Lond) 2025; 49:93-100. [PMID: 39251767 DOI: 10.1038/s41366-024-01625-0] [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] [Received: 03/06/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The study aimed to evaluate how maternal pre-pregnant body mass index (BMI) impacts participant recruitment and retention. METHODS Participants were enrolled in a longitudinal study between 30 and 36 weeks of pregnancy as having normal weight (pre-pregnant BMI ≥ 18.5 and <25 kg/m2) or obesity (pre-pregnant BMI ≥ 30.0 kg/m2). Recruitment channels included Facebook, email, newspaper, phone calls, radio advertisements, flyers, and word-of-mouth. The stages of recruitment included eligibility, consent, and completion. Pearson's chi-square tests were used to evaluate the relationship between BMI and enrollment outcomes. RESULTS Recruitment yielded 2770 total prospective participants. After screening, 141 individuals were eligible, 83 consented, and 60 completed the study. Facebook was the most successful method for identifying eligible pregnant patients with obesity, while a higher percentage of participants recruited through word-of-mouth and flyers consented to the study. Pre-pregnant BMI was significantly associated with the stage of recruitment completed by the participant (p = 0.04), whereby individuals eligible for the study with obesity were less likely to consent and complete study visits. CONCLUSION We demonstrated that maternal obesity was significantly associated with enrollment outcomes in a longitudinal birth cohort study. This study showed that pre-pregnancy BMI influenced study participation. Therefore, tailored recruitment strategies to enhance the recruitment and enrollment of individuals with obesity in maternal-infant health research may be necessary.
Collapse
Affiliation(s)
- Faith Anne N Heeren
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michele Himadi
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Elizabeth Flood-Grady
- STEM Translational Communication Center, University of Florida, Gainesville, FL, USA
- Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Ke Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Matthew Shane Loop
- Department of Health Outcomes Research and Policy, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Magda Francois
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Adetola F Louis-Jacques
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| | - Lindsay Thompson
- Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michelle I Cardel
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
- WW International Inc., New York, NY, USA
| | - Yasmine Gillespie
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lewis DeCicco
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Dominick J Lemas
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL, USA.
- Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA.
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Sugino KY, Janssen RC, McMahan RH, Zimmerman C, Friedman JE, Jonscher KR. Vertical Transfer of Maternal Gut Microbes to Offspring of Western Diet-Fed Dams Drives Reduced Levels of Tryptophan Metabolites and Postnatal Innate Immune Response. Nutrients 2024; 16:1808. [PMID: 38931163 PMCID: PMC11206590 DOI: 10.3390/nu16121808] [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: 05/09/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Maternal obesity and/or Western diet (WD) is associated with an increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in offspring, driven, in part, by the dysregulation of the early life microbiome. Here, using a mouse model of WD-induced maternal obesity, we demonstrate that exposure to a disordered microbiome from WD-fed dams suppressed circulating levels of endogenous ligands of the aryl hydrocarbon receptor (AHR; indole, indole-3-acetate) and TMAO (a product of AHR-mediated transcription), as well as hepatic expression of Il10 (an AHR target), in offspring at 3 weeks of age. This signature was recapitulated by fecal microbial transfer from WD-fed pregnant dams to chow-fed germ-free (GF) lactating dams following parturition and was associated with a reduced abundance of Lactobacillus in GF offspring. Further, the expression of Il10 was downregulated in liver myeloid cells and in LPS-stimulated bone marrow-derived macrophages (BMDM) in adult offspring, suggestive of a hypo-responsive, or tolerant, innate immune response. BMDMs from adult mice lacking AHR in macrophages exhibited a similar tolerogenic response, including diminished expression of Il10. Overall, our study shows that exposure to maternal WD alters microbial metabolites in the offspring that affect AHR signaling, potentially contributing to innate immune hypo-responsiveness and progression of MASLD, highlighting the impact of early life gut dysbiosis on offspring metabolism. Further investigations are warranted to elucidate the complex interplay between maternal diet, gut microbial function, and the development of neonatal innate immune tolerance and potential therapeutic interventions targeting these pathways.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Chelsea Zimmerman
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Li X, Zhu W, Liu R, Ding G, Huang H. Cerium Oxide Nanozymes Improve Skeletal Muscle Function in Gestational Diabetic Offspring by Attenuating Mitochondrial Oxidative Stress. ACS OMEGA 2024; 9:21851-21863. [PMID: 38799328 PMCID: PMC11112706 DOI: 10.1021/acsomega.3c09025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
Gestational diabetes mellitus (GDM) is a significant complication during pregnancy that results in abnormalities in the function of multiple systems in the offspring, which include skeletal muscle dysfunction and reduced systemic metabolic capacity. One of the primary causes behind this intergenerational effect is the presence of mitochondrial dysfunction and oxidative stress in the skeletal muscle of the offspring due to exposure to a high-glucose environment in utero. Cerium oxide (CeO2) nanozymes are antioxidant agents with polymerase activity that have been widely used in the treatment of inflammatory and aging diseases. In this study, we synthesized ultrasmall particle size CeO2 nanozymes and applied them in GDM mouse offspring. The CeO2 nanozymes demonstrated an ability to increase insulin sensitivity and enhance skeletal muscle motility in GDM offspring by improving mitochondrial activity, increasing mitochondrial ATP synthesis function, and restoring abnormal mitochondrial morphology. Furthermore, at the cellular level, CeO2 nanozymes could ameliorate metabolic dysregulation and decrease cell differentiation in adult muscle cells induced by hyperglycemic stimuli. This was achieved through the elimination of endogenous reactive oxygen species (ROS) and an improvement in mitochondrial oxidative respiration function. In conclusion, CeO2 nanozymes play a crucial role in preserving muscle function and maintaining the metabolic stability of organisms. Consequently, they serve to reverse the negative effects of GDM on skeletal muscle physiology in the offspring.
Collapse
Affiliation(s)
- Xinyuan Li
- Obstetrics
and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai200433,China
- Research
Units of Embryo Original Diseases, Chinese
Academy of Medical Sciences (No. 2019RU056), Shanghai200011,China
- Key
Laboratory of Reproductive Genetics (Ministry of Education), Department
of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou310058,China
| | - Wanbo Zhu
- Department
of Orthopedics, Shanghai Sixth People’s Hospital Affiliated
to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Rui Liu
- Obstetrics
and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai200433,China
- Research
Units of Embryo Original Diseases, Chinese
Academy of Medical Sciences (No. 2019RU056), Shanghai200011,China
- Key
Laboratory of Reproductive Genetics (Ministry of Education), Department
of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou310058,China
- Reproductive
Medicine Center, International Institutes of Medicine, the Fourth
Affiliated Hospital, Zhejiang University
School of Medicine, Yiwu322000, China
| | - Guolian Ding
- Obstetrics
and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai200433,China
- Research
Units of Embryo Original Diseases, Chinese
Academy of Medical Sciences (No. 2019RU056), Shanghai200011,China
| | - Hefeng Huang
- Obstetrics
and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai200433,China
- Research
Units of Embryo Original Diseases, Chinese
Academy of Medical Sciences (No. 2019RU056), Shanghai200011,China
- Key
Laboratory of Reproductive Genetics (Ministry of Education), Department
of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou310058,China
| |
Collapse
|
7
|
O'Sullivan A, Brady E, Lafferty L, O'Shea F, O'Regan Z, Meurs N, Baldini M, Gengatharan J, Metallo CM, Wallace M. Long chain monomethyl branched-chain fatty acid levels in human milk vary with gestational weight gain. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102607. [PMID: 38277883 DOI: 10.1016/j.plefa.2024.102607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Breastfeeding is an important determinant of infant health and there is immense interest in understanding its metabolite composition so that key beneficial components can be identified. The aim of this research was to measure the fatty acid composition of human milk in an Irish cohort where we examined changes depending on lactation stage and gestational weight gain trajectory. Utilizing a chromatography approach optimal for isomer separation, we identified 44 individual fatty acid species via GCMS and showed that monomethyl branched-chain fatty acids(mmBCFA's), C15:0 and C16:1 are lower in women with excess gestational weight gain versus low gestational weight gain. To further explore the potential contribution of the activity of endogenous metabolic pathways to levels of these fatty acids in milk, we administered D2O to C57BL/6J dams fed a purified lard based high fat diet (HFD) or low-fat diet during gestation and quantified the total and de novo synthesized levels of fatty acids in their milk. We found that de novo synthesis over three days can account for between 10 and 50 % of mmBCFAs in milk from dams on the low-fat diet dependent on the branched-chain fatty acid species. However, HFD fed mice had significantly decreased de novo synthesized fatty acids in milk resulting in lower total mmBCFAs and medium chain fatty acid levels. Overall, our findings highlight the diverse fatty acid composition of human milk and that human milk mmBCFA levels differ between gestational weight gain phenotypes. In addition, our data indicates that de novo synthesis contributes to mmBCFA levels in mice milk and thus may also be a contributory factor to mmBCFA levels in human milk. Given emerging data indicating mmBCFAs may be beneficial components of milk, this study contributes to our knowledge around the phenotypic factors that may impact their levels.
Collapse
Affiliation(s)
- Aifric O'Sullivan
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Emer Brady
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Lucy Lafferty
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Fiona O'Shea
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Zoe O'Regan
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland
| | - Noah Meurs
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Michelle Baldini
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Jivani Gengatharan
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, CA, USA
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, Salk Institute, 10010N. Torrey Pines Rd., La Jolla, 92037, CA, USA
| | - Martina Wallace
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8, Dublin, Ireland.
| |
Collapse
|
8
|
Alves-Silva T, Húngaro TG, Freitas-Lima LC, de Melo Arthur G, Arruda AC, Santos RB, Oyama LM, Mori MA, Bader M, Araujo RC. Kinin B1 receptor controls maternal adiponectin levels and influences offspring weight gain. iScience 2023; 26:108409. [PMID: 38058311 PMCID: PMC10696114 DOI: 10.1016/j.isci.2023.108409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/16/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Given the importance of the kinin B1 receptor in insulin and leptin hormonal regulation, which in turn is crucial in maternal adaptations to ensure nutrient supply to the fetus, we investigated the role of this receptor in maternal metabolism and fetoplacental development. Wild-type and kinin B1 receptor-deficient (B1KO) female mice were mated with male mice of the opposite genotype. Consequently, the entire litter was heterozygous for kinin B1 receptor, ensuring that there would be no influence of offspring genotype on the maternal phenotype. Maternal kinin B1 receptor blockade reduces adiponectin secretion by adipose tissue ex vivo, consistent with lower adiponectin levels in pregnant B1KO mice. Furthermore, fasting insulinemia also increased, which was associated with placental insulin resistance, reduced placental glycogen accumulation, and heavier offspring. Therefore, we propose the combination of chronic hyperinsulinemia and reduced adiponectin secretion in B1KO female mice create a maternal obesogenic environment that results in heavier pups.
Collapse
Affiliation(s)
- Thaís Alves-Silva
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
- Max-Delbrück Center for Molecular Medicine (MDC), Campus Berlin-Buch, 13125 Berlin, Germany
| | - Talita G.R. Húngaro
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Leandro C. Freitas-Lima
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Gabriel de Melo Arthur
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Adriano C. Arruda
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Raisa B. Santos
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Lila M. Oyama
- Laboratory of Nutrition and Endocrine Physiology, Physiology Department, Federal University of São Paulo (UNIFESP), São Paulo 04023-901, Brazil
| | - Marcelo A.S. Mori
- Laboratory of Aging Biology, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo 13083-862, Brazil
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Campus Berlin-Buch, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité University Medicine Berlin, Berlin, Germany
| | - Ronaldo C. Araujo
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| |
Collapse
|
9
|
Bertin B, Foligne B, Ley D, Lesage J, Beghin L, Morcel J, Gottrand F, Hermann E. An Overview of the Influence of Breastfeeding on the Development of Inflammatory Bowel Disease. Nutrients 2023; 15:5103. [PMID: 38140362 PMCID: PMC10745409 DOI: 10.3390/nu15245103] [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: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The first 1000 days of life is a critical period that contributes significantly to the programming of an individual's future health. Among the many changes that occur during this period early in life, there is growing evidence that the establishment of healthy gut microbiota plays an important role in the prevention of both short- and long-term health problems. Numerous publications suggest that the quality of the gut microbiota colonisation depends on several dietary factors, including breastfeeding. In this respect, a relationship between breastfeeding and the risk of inflammatory bowel disease (IBD) has been suggested. IBDs are chronic intestinal diseases, and perinatal factors may be partly responsible for their onset. We review the existence of links between breastfeeding and IBD based on experimental and clinical studies. Overall, despite encouraging experimental data in rodents, the association between breastfeeding and the development of IBD remains controversial in humans, partly due to the considerable heterogeneity between clinical studies. The duration of exclusive breastfeeding is probably decisive for its lasting effect on IBD. Thus, specific improvements in our knowledge could support dietary interventions targeting the gut microbiome, such as the early use of prebiotics, probiotics or postbiotics, in order to prevent the disease.
Collapse
Affiliation(s)
- Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Benoit Foligne
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| |
Collapse
|
10
|
Ruebel ML, Borengasser SJ, Zhong Y, Kang P, Faske J, Shankar K. Maternal Exercise Prior to and during Gestation Induces Sex-Specific Alterations in the Mouse Placenta. Int J Mol Sci 2023; 24:16441. [PMID: 38003633 PMCID: PMC10671464 DOI: 10.3390/ijms242216441] [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: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
While exercise (EX) during pregnancy is beneficial for both mother and child, little is known about the mechanisms by which maternal exercise mediates changes in utero. Six-week-old female C57BL/6 mice were divided into two groups: with (exercise, EX; N = 7) or without (sedentary, SED; N = 8) access to voluntary running wheels. EX was provided via 24 h access to wheels for 10 weeks prior to conception until late pregnancy (18.5 days post coitum). Sex-stratified placentas and fetal livers were collected. Microarray analysis of SED and EX placentas revealed that EX affected gene transcript expression of 283 and 661 transcripts in male and female placentas, respectively (±1.4-fold, p < 0.05). Gene Set Enrichment and Ingenuity Pathway Analyses of male placentas showed that EX led to inhibition of signaling pathways, biological functions, and down-regulation of transcripts related to lipid and steroid metabolism, while EX in female placentas led to activation of pathways, biological functions, and gene expression related to muscle growth, brain, vascular development, and growth factors. Overall, our results suggest that the effects of maternal EX on the placenta and presumably on the offspring are sexually dimorphic.
Collapse
Affiliation(s)
- Meghan L. Ruebel
- Microbiome and Metabolism Research Unit, USDA-ARS, Southeast Area, Little Rock, AR 72202, USA;
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Sarah J. Borengasser
- Tobacco Settlement Endowment Trust Health Promotion Research Center, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics—Endocrinology & Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ying Zhong
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Ping Kang
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Jennifer Faske
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
11
|
Calderari S, Archilla C, Jouneau L, Daniel N, Peynot N, Dahirel M, Richard C, Mourier E, Schmaltz-Panneau B, Vitorino Carvalho A, Rousseau-Ralliard D, Lager F, Marchiol C, Renault G, Gatien J, Nadal-Desbarats L, Couturier-Tarrade A, Duranthon V, Chavatte-Palmer P. Alteration of the embryonic microenvironment and sex-specific responses of the preimplantation embryo related to a maternal high-fat diet in the rabbit model. J Dev Orig Health Dis 2023; 14:602-613. [PMID: 37822211 DOI: 10.1017/s2040174423000260] [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: 10/13/2023]
Abstract
The maternal metabolic environment can be detrimental to the health of the offspring. In a previous work, we showed that maternal high-fat (HH) feeding in rabbit induced sex-dependent metabolic adaptation in the fetus and led to metabolic syndrome in adult offspring. As early development representing a critical window of susceptibility, in the present work we aimed to explore the effects of the HH diet on the oocyte, preimplantation embryo and its microenvironment. In oocytes from females on HH diet, transcriptomic analysis revealed a weak modification in the content of transcripts mainly involved in meiosis and translational control. The effect of maternal HH diet on the embryonic microenvironment was investigated by identifying the metabolite composition of uterine and embryonic fluids collected in vivo by biomicroscopy. Metabolomic analysis revealed differences in the HH uterine fluid surrounding the embryo, with increased pyruvate concentration. Within the blastocoelic fluid, metabolomic profiles showed decreased glucose and alanine concentrations. In addition, the blastocyst transcriptome showed under-expression of genes and pathways involved in lipid, glucose and amino acid transport and metabolism, most pronounced in female embryos. This work demonstrates that the maternal HH diet disrupts the in vivo composition of the embryonic microenvironment, where the presence of nutrients is increased. In contrast to this nutrient-rich environment, the embryo presents a decrease in nutrient sensing and metabolism suggesting a potential protective process. In addition, this work identifies a very early sex-specific response to the maternal HH diet, from the blastocyst stage.
Collapse
Affiliation(s)
- Sophie Calderari
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Catherine Archilla
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Nathalie Daniel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Nathalie Peynot
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Michele Dahirel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Christophe Richard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
- Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Eve Mourier
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
- Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Barbara Schmaltz-Panneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Anaïs Vitorino Carvalho
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Delphine Rousseau-Ralliard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Franck Lager
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Carmen Marchiol
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Gilles Renault
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Julie Gatien
- Research and Development Department, Eliance, Nouzilly, France
| | - Lydie Nadal-Desbarats
- UMR 1253, iBrain, University of Tours, Inserm, Tours, France
- PST-ASB, University of Tours, Tours, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Véronique Duranthon
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| |
Collapse
|
12
|
Peterson AK, Zhu Y, Fuller S, Feng J, Alexeeff S, Mitro SD, Kannan K, Robinson M, Padula A, Ferrara A. PFAS concentrations in early and mid-pregnancy and risk of gestational diabetes mellitus in a nested case-control study within the ethnically and racially diverse PETALS cohort. BMC Pregnancy Childbirth 2023; 23:657. [PMID: 37704943 PMCID: PMC10500777 DOI: 10.1186/s12884-023-05953-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are persistent synthetic chemicals and are commonly found in everyday items. PFAS have been linked to disrupting glucose homeostasis, however, whether they are associated with gestational diabetes mellitus (GDM) risk remains inconclusive. We examined prospective associations of PFAS concentrations measured twice in pregnancy with GDM risk. METHODS In the PETALS pregnancy cohort, a nested case-control study which included 41 GDM cases and 87 controls was conducted. PFAS analytes were measured in blood serum collected in both early and mid-pregnancy (mean [SD]: 13.9 [2.2] and 20.2 [2.2] gestational weeks, respectively), with cumulative exposure calculated by the area-under-the-curve (AUC) to integrate both the PFAS concentration and the timing of the exposure. Individual adjusted weighted unconditional logistic regression models examined seven PFAS in association with GDM risk. P-values were corrected using the false-discovery-rate (FDR). Mixture models were analyzed with Bayesian kernel machine regression (BKMR). RESULTS PFDA, PFNA and PFOA were individually associated with higher GDM risk per interquartile range (IQR) in early pregnancy (OR [95% CI]: 1.23 [1.09, 1.38]), 1.40 [1.24, 1.58]), and 1.15 [1.04, 1.27], respectively), mid-pregnancy (1.28 [1.15, 1.43], 1.16 [1.05, 1.28], and 1.20 [1.09, 1.33], respectively), and with cumulative exposure (1.23 [1.09, 1.38], 1.21 [1.07, 1.37], and 1.19 [1.09, 1.31], respectively). PFOS in mid-pregnancy and with cumulative exposure was associated with increased GDM risk (1.41 [1.17, 1.71] and 1.33 [1.06, 1.58], respectively). PFUnDA in early pregnancy was associated with lower GDM risk (0.79 [0.64, 0.98]), whereas mid-pregnancy levels were associated with higher risk (1.49 [1.18, 1.89]). PFHxS was associated with decreased GDM risk in early and mid-pregnancy (0.48 [0.38, 0.60] and 0.48 [0.37, 0.63], respectively) and with cumulative exposure (0.49 [0.38,0.63]). PFPeA was not associated with GDM. Similar conclusions were observed in BKMR models; however, overall associations in these models were not statistically significant. CONCLUSIONS Higher risk of GDM was consistently observed in association with PFDA, PFNA, and PFOA exposure in both early and mid-pregnancy. Results should be corroborated in larger population-based cohorts and individuals of reproductive age should potentially avoid known sources of PFAS.
Collapse
Affiliation(s)
- Alicia K Peterson
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA.
- Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA.
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
- Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Sophia Fuller
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Juanran Feng
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Susanna D Mitro
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
- Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Kurunthachalam Kannan
- Department of Pediatrics, New York University School of Medicine, 145 East 32 Street, New York, NY, 10016, USA
| | - Morgan Robinson
- Department of Pediatrics, New York University School of Medicine, 145 East 32 Street, New York, NY, 10016, USA
| | - Amy Padula
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, 490 Illinois Street, San Francisco, 94143 CA, USA
| | - Assiamira Ferrara
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
- Center for Upstream Prevention of Adiposity and Diabetes Mellitus (UPSTREAM), Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| |
Collapse
|
13
|
Gaillard R, Jaddoe VWV. Maternal cardiovascular disorders before and during pregnancy and offspring cardiovascular risk across the life course. Nat Rev Cardiol 2023; 20:617-630. [PMID: 37169830 DOI: 10.1038/s41569-023-00869-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/13/2023]
Abstract
Obesity, hypertension, type 2 diabetes mellitus and dyslipidaemia are highly prevalent among women of reproductive age and contribute to complications in >30% of pregnancies in Western countries. An accumulating body of evidence suggests that these cardiovascular disorders in women, occurring before and during their pregnancy, can affect the development of the structure, physiology and function of cardiovascular organ systems at different stages during embryonic and fetal development. These developmental adaptations might, in addition to genetics and sociodemographic and lifestyle factors, increase the susceptibility of the offspring to cardiovascular disease throughout the life course. In this Review, we discuss current knowledge of the influence of maternal cardiovascular disorders, occurring before and during pregnancy, on offspring cardiovascular development, dysfunction and disease from embryonic life until adulthood. We discuss findings from contemporary, large-scale, observational studies that provide insights into specific critical periods, evidence for causality and potential underlying mechanisms. Furthermore, we focus on priorities for future research, including defining optimal cardiovascular and reproductive health in women and men before their pregnancy and identifying specific embryonic, placental and fetal molecular developmental adaptations from early pregnancy onwards. Together, these approaches will help stop the intergenerational cycle of cardiovascular disease.
Collapse
Affiliation(s)
- Romy Gaillard
- Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, Netherlands.
| | - Vincent W V Jaddoe
- Department of Paediatrics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
14
|
Nagagata BA, Ajackson M, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Obese mothers supplemented with melatonin during gestation and lactation ameliorate the male offspring's pancreatic islet cellular composition and beta-cell function. J Dev Orig Health Dis 2023; 14:490-500. [PMID: 37366144 DOI: 10.1017/s2040174423000168] [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: 06/28/2023]
Abstract
Melatonin supplementation to obese mothers during gestation and lactation might benefit the pancreatic islet cellular composition and beta-cell function in male offspring adulthood. C57BL/6 females (mothers) were assigned to two groups (n = 20/each) based on their consumption in control (C 17% kJ as fat) or high-fat diet (HF 49% kJ as fat). Mothers were supplemented with melatonin (Mel) (10 mg/kg daily) during gestation and lactation, or vehicle, forming the groups (n = 10/each): C, CMel, HF, and HFMel. The male offspring were studied, considering they only received the C diet after weaning until three months old. The HF mothers and their offspring showed higher body weight, glucose intolerance, insulin resistance, and low insulin sensitivity than the C ones. However, HFMel mothers and their offspring showed improved glucose metabolism and weight loss than the HF ones. Also, the offspring's higher expressions of pro-inflammatory markers and endoplasmic reticulum (ER) stress were observed in HF but reduced in HFMel. Contrarily, antioxidant enzymes were less expressed in HF but improved in HFMel. In addition, HF showed increased beta-cell mass and hyperinsulinemia but diminished in HFMel. Besides, the beta-cell maturity and identity gene expressions diminished in HF but enhanced in HFMel. In conclusion, obese mothers supplemented with melatonin benefit their offspring's islet cell remodeling and function. In addition, improving pro-inflammatory markers, oxidative stress, and ER stress resulted in better glucose and insulin levels control. Consequently, pancreatic islets and functioning beta cells were preserved in the offspring of obese mothers supplemented with melatonin.
Collapse
Affiliation(s)
- Brenda A Nagagata
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Ajackson
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Harmancıoğlu B, Kabaran S. Maternal high fat diets: impacts on offspring obesity and epigenetic hypothalamic programming. Front Genet 2023; 14:1158089. [PMID: 37252665 PMCID: PMC10211392 DOI: 10.3389/fgene.2023.1158089] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Maternal high-fat diet (HFD) during pregnancy is associated with rapid weight gain and fetal fat mass increase at an early stage. Also, HFD during pregnancy can cause the activation of proinflammatory cytokines. Maternal insulin resistance and inflammation lead to increased adipose tissue lipolysis, and also increased free fatty acid (FFA) intake during pregnancy (˃35% of energy from fat) cause a significant increase in FFA levels in the fetus. However, both maternal insulin resistance and HFD have detrimental effects on adiposity in early life. As a result of these metabolic alterations, excess fetal lipid exposure may affect fetal growth and development. On the other hand, increase in blood lipids and inflammation can adversely affect the development of the liver, adipose tissue, brain, skeletal muscle, and pancreas in the fetus, increasing the risk for metabolic disorders. In addition, maternal HFD is associated with changes in the hypothalamic regulation of body weight and energy homeostasis by altering the expression of the leptin receptor, POMC, and neuropeptide Y in the offspring, as well as altering methylation and gene expression of dopamine and opioid-related genes which cause changes in eating behavior. All these maternal metabolic and epigenetic changes may contribute to the childhood obesity epidemic through fetal metabolic programming. Dietary interventions, such as limiting dietary fat intake <35% with appropriate fatty acid intake during the gestation period are the most effective type of intervention to improve the maternal metabolic environment during pregnancy. Appropriate nutritional intake during pregnancy should be the principal goal in reducing the risks of obesity and metabolic disorders.
Collapse
|
16
|
Mukoma G, Norris SA, Chikowore T. Nutrient Patterns and Body Mass Index: A Comparative Longitudinal Analysis in Urban Black South African Adolescents and Adults. Nutrients 2023; 15:nu15051075. [PMID: 36904075 PMCID: PMC10004796 DOI: 10.3390/nu15051075] [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: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
OBJECTIVE We set out to evaluate the association between nutrient patterns and general adiposity in black South African adolescents and adults and to determine whether the interactions are longitudinally sustained over 24 months. METHODS Principal Component Analysis (PCA) was used to derive the nutrient patterns of 750 participants (250 adolescents between 13 and 17 years old and 500 adults who were 27 years or 45+ years old). PCA was applied to 25 nutrients, computed from the quantified food frequency questionnaire (QFFQ) over a 24 months period. RESULTS The nutrient patterns between adolescents and adults were similar over time; however, their associations with BMI were different. Among the adolescents, only the "plant-driven nutrients pattern" was significantly associated with a 0.56% (95% CI (0.33; 0.78); p < 0.001) increase in BMI. Among the adults, the "plant-driven nutrient pattern" (0.43% (95% CI (0.03; 0.85); p < 0.001) and the "fat-driven nutrients pattern" (0.18% (95% CI (0.06; 0.29); p < 0.001) were significantly associated with a BMI increase. Furthermore, the "plant-driven nutrient pattern", "fat-driven nutrient pattern" and the animal-driven nutrient pattern revealed sex differences in their association with BMI. CONCLUSION Urban adolescents and adults had consistent nutrient patterns, but their BMI relationships changed with age and gender, an important finding for future nutrition interventions.
Collapse
Affiliation(s)
- Gudani Mukoma
- SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 1862, South Africa
- Correspondence: ; Tel.: +27-799037819
| | - Shane A. Norris
- SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 1862, South Africa
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Tinashe Chikowore
- SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 1862, South Africa
| |
Collapse
|
17
|
Cleveland JC, Espinoza J, Holzhausen EA, Goran MI, Alderete TL. The impact of social determinants of health on obesity and diabetes disparities among Latino communities in Southern California. BMC Public Health 2023; 23:37. [PMID: 36609302 PMCID: PMC9817265 DOI: 10.1186/s12889-022-14868-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Social determinants of health (SDoH) describe the complex network of circumstances that impact an individual before birth and across the lifespan. SDoH contextualize factors in a community that are associated with chronic disease risk and certain health disparities. The main objective of this study was to explore the impact of SDoH on the prevalence of obesity and diabetes, and whether these factors explain disparities in these health outcomes among Latinos in Southern California. METHODS We utilized three composite indices that encompass different SDoH: the Healthy Places Index (HPI), Social Vulnerability Index (SVI), and CalEnviroScreen (CES). Univariate linear regression models explored the associations between index scores with adult obesity, adult diabetes, and childhood obesity. RESULTS Communities with lower HPI scores were associated with higher prevalence of metabolic disease and a greater proportion of Latino residents. Cities in the lowest decile of HPI scores had 71% of the population identifying as Latino compared to 12% in the highest decile. HPI scores explained 61% of the variability in adult obesity (p < 0.001), 41% of the variability in childhood obesity (p < 0.001), and 47% of the variability in adult diabetes (p < 0.001). Similar results were observed when examining SVI and CES with these health outcomes. CONCLUSIONS These results suggest that Latinos in Southern California live in communities with adverse SDoH and face a greater burden of adult obesity, diabetes, and childhood obesity.
Collapse
Affiliation(s)
- Joseph C Cleveland
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Juan Espinoza
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | | | - Michael I Goran
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
18
|
Navarrete E, Díaz G, Salazar AM, Montúfar-Chaveznava R, Caldelas I. Long-term changes in the diurnal temporal regulation and set points of metabolic parameters associated with chronic maternal overnutrition in rabbits. Am J Physiol Endocrinol Metab 2022; 323:E503-E516. [PMID: 36288336 DOI: 10.1152/ajpendo.00144.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MS) and obesity have become a worldwide epidemic with an alarming prevalence in women of reproductive age. Maternal metabolic condition is considered a risk factor for adverse birth outcomes and long-term MS. In this study, we developed a rabbit model of maternal overnutrition via the chronic intake of a high-fat and carbohydrate diet (HFCD), and we determined the effects of this diet on maternal metabolism and offspring metabolic set points and temporal metabolic regulation in adult life. Before and during pregnancy, the female rabbits that consumed the HFCD exhibited significant changes in body weight, serum levels of analytes associated with carbohydrate and lipid metabolism, levels of liver and kidney damage markers, and liver histology. Our data suggest that rabbits are a valuable model for studying the development of MS associated with the chronic intake of unbalanced diets and fetal metabolic programming. Furthermore, the offspring of overnourished dams exhibited considerable changes in 24-h serum metabolite profiles in adulthood, with notable sexual dimorphism. These data suggest that maternal nutritional conditions due to the chronic intake of an HFCD adversely impact key elements related to the development of circadian rhythmicity in offspring.NEW & NOTEWORTHY Maternal overnutrition previous and during pregnancy leads to long-term changes in the 24-h regulation and setpoint of metabolic profiles of the offspring.
Collapse
Affiliation(s)
- Erika Navarrete
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Georgina Díaz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ana María Salazar
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | | | - Ivette Caldelas
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
19
|
Cechinel LR, Batabyal RA, Freishtat RJ, Zohn IE. Parental obesity-induced changes in developmental programming. Front Cell Dev Biol 2022; 10:918080. [PMID: 36274855 PMCID: PMC9585252 DOI: 10.3389/fcell.2022.918080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies support the link between parental obesity and the predisposition to develop adult-onset metabolic syndromes that include obesity, high blood pressure, dyslipidemia, insulin resistance, and diabetes in the offspring. As the prevalence of obesity increases in persons of childbearing age, so does metabolic syndrome in their descendants. Understanding how parental obesity alters metabolic programs in the progeny, predisposing them to adult-onset metabolic syndrome, is key to breaking this cycle. This review explores the basis for altered metabolism of offspring exposed to overnutrition by focusing on critical developmental processes influenced by parental obesity. We draw from human and animal model studies, highlighting the adaptations in metabolism that occur during normal pregnancy that become maladaptive with obesity. We describe essential phases of development impacted by parental obesity that contribute to long-term alterations in metabolism in the offspring. These encompass gamete formation, placentation, adipogenesis, pancreas development, and development of brain appetite control circuits. Parental obesity alters the developmental programming of these organs in part by inducing epigenetic changes with long-term consequences on metabolism. While exposure to parental obesity during any of these phases is sufficient to alter long-term metabolism, offspring often experience multiple exposures throughout their development. These insults accumulate to increase further the susceptibility of the offspring to the obesogenic environments of modern society.
Collapse
|
20
|
Perinatal exposure to isocaloric diet with moderate-fat promotes pancreatic islets insulin hypersecretion and susceptibility to islets exhaustion in response to fructose intake in adult male rat offspring. Life Sci 2022; 307:120873. [PMID: 35952730 DOI: 10.1016/j.lfs.2022.120873] [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: 06/06/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 11/21/2022]
Abstract
AIMS Perinatal maternal hypercaloric diets increase the susceptibility to metabolic disorders in the offspring. We hypothesized that maternal intake of an isocaloric moderate-fat diet (mMFD) would disturb the glucose homeostasis and favor the β-cell failure in response to fructose overload in adult male offspring. METHODS Female Wistar rats received an isocaloric diet (3.9 kcal/g) containing 29 % (mMFD) or 9 % as fat (mSTD) prior mating and throughout gestation and lactation. After weaning, male offspring received standard chow and fructose-drinking water (15 %) between 120 and 150 days old. KEY FINDINGS mMFD offspring had higher body weight, visceral adiposity and, fasting glycemia, with normal insulinemia. Fructose increased glycemia at 15 min from oral glucose administration, but only mMFD had returned to basal glucose levels at 120 min. Fructose increased HOMA-IR index regardless diet, but only mMFD exhibited hyperinsulinemia and a higher HOMA-β index. mMFD pancreatic islets showed increased area and insulin immunostaining density, suggesting β-cell hypertrophy. Fructose induced the expected compensatory hypertrophy in mSTD islets, while the opposite occurred in mMFD islets, associated with reduced insulin immunostaining, suggesting lower insulin storage. Pancreatic islets isolated from mMFD offspring exhibited higher glucose-stimulated insulin release at physiological concentrations. However, at higher glucose concentrations, the islets from fructose-treated mMFD reduced dramatically their insulin release, suggesting exhaustion. SIGNIFICANCE Isocaloric mMFD induced adaptive mechanism in the offspring allowing insulin hypersecretion, but under metabolic challenge with fructose, β-cell compensation shifts to exhaustion, favoring dysfunction. Therefore, a maternal MFD may contribute to developing diabetes under fructose overload in the adult offspring.
Collapse
|
21
|
Zhu BB, Gao H, Geng ML, Wu X, Tong J, Deng F, Zhang SY, Wu LH, Huang K, Wu XY, Gan H, Zhu P, Tao FB. Sex Discrepancy Observed for Gestational Metabolic Syndrome Parameters and Polygenic Risk Associated With Preschoolers' BMI Growth Trajectory: The Ma'anshan Birth Cohort Study. Front Endocrinol (Lausanne) 2022; 13:857711. [PMID: 35846310 PMCID: PMC9283700 DOI: 10.3389/fendo.2022.857711] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Few studies have investigated the associations of childhood growth trajectories with the prenatal metabolic risks of mothers and their interaction with children's genetic susceptibility. Objective To investigate the effects of gestational metabolic syndrome (GMS) risks and children's polygenic risk scores (PRSs), and their interaction effect on the BMI trajectory and obesity risk of offspring from birth to 6 years of age. Methods A total of 2,603 mother-child pairs were recruited from the Ma'anshan birth cohort (Anhui Province of China) study. Data on maternal prepregnancy obesity, gestational weight gain (GWG), gestational diabetes mellitus (GDM), and hypertensive disorders of pregnancy (HDP) were used to evaluate maternal GMS risk. In addition, 1,482 cord blood samples were used to genotype 11 candidate single-nucleotide polymorphisms (SNPs) to calculate children's PRSs. The latent class growth model using the longitudinal BMI-for-age z scores (BMIz) was applied to validly capture the BMIz growth trajectory. Results Maternal GMS status was associated with higher BMIz scores and with an increased risk of overweight/obesity. Positive relationships were revealed between PRS and the risk of overweight/obesity among girls. Additionally, maternal GMS significantly interacted with the child's PRS on BMIz scores and the risk of overweight/obesity among girls. Hierarchical BMI trajectory graphs by different exposure groups showed consistent findings, and both boys' and girls' BMIz trajectories were divided into three groups. Among girls, the higher the GMS risk or PRS they had, the higher the probability of being in the high BMIz trajectory group. Conclusions Maternal GMS status increased BMIz scores and the risk of obesity in both boys and girls and elevated the child's BMI trajectory from birth to 6 years of age among girls. PRSs were significantly associated with children's BMI trajectory and the risk of obesity and modified the associations between maternal GMS status and obesity biomarkers only among girls. Thus, regarding childhood obesity, steps should be taken to decrease maternal metabolic risks before and during pregnancy, and sex discrepancies should be noted to identify high-risk populations after birth to hierarchically manage them.
Collapse
Affiliation(s)
- Bei-bei Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Hui Gao
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, (Anhui Medical University), National Health Commission of the People’s Republic China, Hefei, China
| | - Meng-long Geng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Xiulong Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Fen Deng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Si-ying Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Li-hong Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Xiao-yan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Peng Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Fang-biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| |
Collapse
|
22
|
Wiedmer EB, Herter-Aeberli I. The Potential of Prebiotic and Probiotic Supplementation During Obese Pregnancy to Improve Maternal and Offspring’s Metabolic Health and Reduce Obesity Risk—A Narrative Review. Front Nutr 2022; 9:819882. [PMID: 35464026 PMCID: PMC9021550 DOI: 10.3389/fnut.2022.819882] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/14/2022] [Indexed: 01/14/2023] Open
Abstract
Worldwide, obesity prevalence is rising, severely impairing the health of those affected by increasing their risk for developing non-communicable diseases. The pathophysiology of obesity is complex and caused by a variety of genetic and environmental factors. Recent findings suggest that obesity is partly caused by dysbiosis, an imbalanced gut microbiome. In the context of pregnancy, maternal dysbiosis increases the child’s obesity risk, causing an intergenerational cycle of obesity. Accordingly, interventions modulating the gut microbiome have the potential to interrupt this cycle. This review discusses the potential of pre- and probiotic interventions in modulating maternal obesity associated dysbiosis to limit the child’s obesity risk. The literature search resulted in four animal studies using prebiotics as well as one animal study and six human studies using probiotics. Altogether, prebiotic supplementation in animals successfully decreased the offspring’s obesity risk, while probiotic supplementation in humans failed to show positive impacts in the offspring. However, comparability between studies is limited and considering the complexity of the topic, more studies in this field are required.
Collapse
|
23
|
Mennitti LV, Carpenter AAM, Loche E, Pantaleão LC, Fernandez-Twinn DS, Schoonejans JM, Blackmore HL, Ashmore TJ, Pisani LP, Tadross JA, Hargreaves I, Ozanne SE. Effects of maternal diet-induced obesity on metabolic disorders and age-associated miRNA expression in the liver of male mouse offspring. Int J Obes (Lond) 2022; 46:269-278. [PMID: 34663892 PMCID: PMC8794789 DOI: 10.1038/s41366-021-00985-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/22/2021] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study investigated the effect of maternal obesity on aged-male offspring liver phenotype and hepatic expression of a programmed miRNA. METHODS A mouse model (C57BL/6 J) of maternal diet-induced obesity was used to investigate fasting-serum metabolites, hepatic lipid content, steatosis, and relative mRNA levels (RT-PCR) and protein expression (Western blotting) of key components involved in hepatic and mitochondrial metabolism in 12-month-old offspring. We also measured hepatic lipid peroxidation, mitochondrial content, fibrosis stage, and apoptosis in the offspring. To investigate potential mechanisms leading to the observed phenotype, we also measured the expression of miR-582 (a miRNA previously implicated in liver cirrhosis) in 8-week-old and 12-month-old offspring. RESULTS Body weight and composition was similar between 8-week-old offspring, however, 12-month-old offspring from obese mothers had increased body weight and fat mass (19.5 ± 0.8 g versus 10.4 ± 0.9 g, p < 0.001), as well as elevated serum levels of LDL and leptin and hepatic lipid content (21.4 ± 2.1 g versus 12.9 ± 1.8 g, p < 0.01). This was accompanied by steatosis, increased Bax/Bcl-2 ratio, and overexpression of p-SAPK/JNK, Tgfβ1, Map3k14, and Col1a1 in the liver. Decreased levels of Bcl-2, p-AMPKα, total AMPKα and mitochondrial complexes were also observed. Maternal obesity was associated with increased hepatic miR-582-3p (p < 0.001) and miR-582-5p (p < 0.05). Age was also associated with an increase in both miR-582-3p and miR-582-5p, however, this was more pronounced in the offspring of obese dams, such that differences were greater in 12-month-old animals (-3p: 7.34 ± 1.35 versus 1.39 ± 0.50, p < 0.0001 and -5p: 4.66 ± 1.16 versus 1.63 ± 0.65, p < 0.05). CONCLUSION Our findings demonstrate that maternal diet-induced obesity has detrimental effects on offspring body composition as well as hepatic phenotype that may be indicative of accelerated-ageing phenotype. These whole-body and cellular phenotypes were associated with age-dependent changes in expression of miRNA-582 that might contribute mechanistically to the development of metabolic disorders in the older progeny.
Collapse
Affiliation(s)
- Laís Vales Mennitti
- Department of Bioscience, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, 11015-020, Brazil
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Asha A M Carpenter
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Elena Loche
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Lucas C Pantaleão
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Josca M Schoonejans
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Heather L Blackmore
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Thomas J Ashmore
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - Luciana Pellegrini Pisani
- Department of Bioscience, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, 11015-020, Brazil
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
| | - John A Tadross
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | - Iain Hargreaves
- Department of Pharmacy and Biomolecular Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 5UA, United Kingdom
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, CB2 OQQ, United Kingdom.
| |
Collapse
|
24
|
Chen J, Yang X, Huang L, Zhang Z, Yao J, Liang H, Zhou W. Insulin resistance biomarkers in small-for-gestational-age infants born to mothers with gestational diabetes mellitus. J Matern Fetal Neonatal Med 2021; 35:9061-9065. [PMID: 34913819 DOI: 10.1080/14767058.2021.2014449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Early alterations in glucose homeostasis increase the risk of developing insulin resistance (IR) and obesity later in life. The study aimed to ascertain the peripheral blood levels of hormones that controlling glucose homeostasis and inflammatory factors that are correlated with IR and fetal outcomes in small-for-gestational-age (SGA) infants born to mothers with gestational diabetes mellitus (GDM). METHODS This cohort study included a total of 90 SGA infants born to mothers with GDM (n = 37) and without GDM (n = 53). At birth, blood levels of glucose, insulin, C-peptide, growth hormone (GH), IGFBP3, lipid profiles, fibrinogen, and hypersensitive C-reactive protein (Hs-CRP) were measured; homeostatic model assessment-IR (HOMA-IR) and ponderal index were calculated. All newborns were followed up to the first year of life. RESULTS Compared with SGA infants born to mothers without GDM, the levels of low-density lipoprotein-cholesterol (LDL-C), GH, and fibrinogen were significantly higher in the SGA infants born to mothers with GDM (p = .048, .045, and .04, respectively). However, total cholesterol, HDL-C, and apolipoprotein(a) levels were significantly lower in the SGA infants born to mothers with GDM when compared with those in with SGA infants born to mothers without GDM (all p < .05). Weight gain in the first year was higher in the SGA infants born to mothers with GDM group than SGA infants born to mothers without GDM [6644 g (5991-7572) vs. 6032 g (5529-6932)]. CONCLUSIONS Altered biomarkers of IR were observed among SGA infants born to mothers with GDM, suggesting that these infants were more prone to develop IR after birth.
Collapse
Affiliation(s)
- Juncao Chen
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Xiaoqi Yang
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Longguang Huang
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Zhe Zhang
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Jie Yao
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Huiying Liang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
25
|
Ramalingam L, Menikdiwela KR, Spainhour S, Eboh T, Moustaid-Moussa N. Sex Differences in Early Programming by Maternal High Fat Diet Induced-Obesity and Fish Oil Supplementation in Mice. Nutrients 2021; 13:3703. [PMID: 34835957 PMCID: PMC8625698 DOI: 10.3390/nu13113703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022] Open
Abstract
Pre-pregnancy obesity is a contributing factor for impairments in offspring metabolic health. Interventional strategies during pregnancy are a potential approach to alleviate and/or prevent obesity and obesity related metabolic alterations in the offspring. Fish oil (FO), rich in omega-3 polyunsaturated fatty acids (n-3 PUFAs) exerts metabolic health benefits. However, the role of FO in early life remains still unknown. Hence, this study objective was to determine the effect of FO supplementation in mice from pre-pregnancy through lactation, and to study the post-natal metabolic health effects in gonadal fat and liver of offspring fed high fat (HF) diet with or without FO. Female C57BL6J mice aged 4-5 weeks were fed a HF (45% fat) diet supplemented with or without FO (30 g/kg of diet) and low fat (LF; 10% fat) pre-pregnancy through lactation. After weaning, offspring (male and female) from HF or FO dams either continued the same diet (HF-HF and FO-FO) or switched to the other diet (HF-FO and FO-HF) for 13 weeks, creating four groups of treatment, and LF-LF was used as a control group. Serum, gonadal fat and liver tissue were collected at termination for metabolic analyses. Offspring of both sexes fed HF with or without fish oil gained (p < 0.05) more weight post weaning, compared to LF-LF-fed mice. All the female offspring groups supplemented with FO had reduced body weight compared to the respective male groups. Further, FO-FO supplementation in both sexes (p < 0.05) improved glucose clearance and insulin sensitivity compared to HF-HF. All FO-FO fed mice had significantly reduced adipocyte size compared to HF-HF group in both male and females. Inflammation, measured by mRNA levels of monocyte chemoattractant protein 1 (Mcp1), was reduced (p < 0.05) with FO supplementation in both sexes in gonadal fat and in the liver. Markers of fatty acid synthesis, fatty acid synthase (Fasn) showed no sex specific differences in gonadal fat and liver of mice supplemented with HF. Female mice had lower liver triglycerides than male counterparts. Supplementation of FO in mice improved metabolic health of offspring by lowering markers of lipid synthesis and inflammation.
Collapse
Affiliation(s)
- Latha Ramalingam
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX 74909, USA; (L.R.); (K.R.M.); (S.S.); (T.E.)
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY 13244, USA
| | - Kalhara R. Menikdiwela
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX 74909, USA; (L.R.); (K.R.M.); (S.S.); (T.E.)
| | - Stephani Spainhour
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX 74909, USA; (L.R.); (K.R.M.); (S.S.); (T.E.)
| | - Tochi Eboh
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX 74909, USA; (L.R.); (K.R.M.); (S.S.); (T.E.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX 74909, USA; (L.R.); (K.R.M.); (S.S.); (T.E.)
| |
Collapse
|
26
|
César H, Sertorio MN, de Souza EA, Jamar G, Santamarina A, Jucá A, Casagrande BP, Pisani LP. Parental high-fat high-sugar diet programming and hypothalamus adipose tissue axis in male Wistar rats. Eur J Nutr 2021; 61:523-537. [PMID: 34657184 DOI: 10.1007/s00394-021-02690-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/28/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE Maternal nutrition during early development and paternal nutrition pre-conception can programme offspring health status. Hypothalamus adipose axis is a target of developmental programming, and paternal and maternal high-fat, high-sugar diet (HFS) may be an important factor that predisposes offspring to develop obesity later in life. This study aims to investigate Wistar rats' maternal and paternal HFS differential contribution on the development, adiposity, and hypothalamic inflammation in male offspring from weaning until adulthood. METHODS Male progenitors were fed a control diet (CD) or HFS for 10 weeks before mating. After mating, dams were fed CD or HFS only during pregnancy and lactation. Forming the following male offspring groups: CD-maternal and paternal CD; MH-maternal HFS and paternal CD; PH-maternal CD and paternal HFS; PMH-maternal and paternal HFS. After weaning, male offspring were fed CD until adulthood. RESULTS Maternal HFS diet increased weight, visceral adiposity, and serum total cholesterol levels, and decreased hypothalamic weight in weanling male rats. In adult male offspring, maternal HFS increased weight, glucose levels, and hypothalamic NFκBp65. Paternal HFS diet lowered hypothalamic insulin receptor levels in weanling offspring and glucose and insulin levels in adult offspring. The combined effects of maternal and paternal HFS diets increased triacylglycerol, leptin levels, and hypothalamic inflammation in weanling rats, and increased visceral adiposity in adulthood. CONCLUSION Male offspring intake of CD diet after weaning reversed part of the effects of parental HFS diet during the perinatal period. However, maternal and paternal HFS diet affected adiposity and hypothalamic inflammation, which remained until adulthood.
Collapse
Affiliation(s)
- Helena César
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo-UNIFESP, Santos, SP, Brazil
| | | | - Esther Alves de Souza
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Giovana Jamar
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Aline Santamarina
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Andrea Jucá
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Breno Picin Casagrande
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
27
|
Understanding the Long-Lasting Effects of Fetal Nutrient Restriction versus Exposure to an Obesogenic Diet on Islet-Cell Mass and Function. Metabolites 2021; 11:metabo11080514. [PMID: 34436455 PMCID: PMC8401811 DOI: 10.3390/metabo11080514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/14/2022] Open
Abstract
Early life represents a window of phenotypic plasticity. Thus, exposure of the developing fetus to a compromised nutritional environment can have long term consequences for their health. Indeed, undernutrition or maternal intake of an obesogenic diet during pregnancy leads to a heightened risk of type 2 diabetes (T2D) and obesity in her offspring in adult life. Given that abnormalities in beta-cell function are crucial in delineating the risk of T2D, studies have investigated the impact of these exposures on islet morphology and beta-cell function in the offspring in a bid to understand why they are more at risk of T2D. Interestingly, despite the contrasting maternal metabolic phenotype and, therefore, intrauterine environment associated with undernutrition versus high-fat feeding, there are a number of similarities in the genes/biological pathways that are disrupted in offspring islets leading to changes in function. Looking to the future, it will be important to define the exact mechanisms involved in mediating changes in the gene expression landscape in islet cells to determine whether the road to T2D development is the same or different in those exposed to different ends of the nutritional spectrum.
Collapse
|
28
|
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
|
29
|
Postnatally induced metabolic and oxidative changes associated with maternal high-fat consumption were mildly affected by Quercetin-3-O-rutinoside treatment in rats. Heliyon 2021; 7:e06847. [PMID: 33997389 PMCID: PMC8102762 DOI: 10.1016/j.heliyon.2021.e06847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/05/2021] [Accepted: 04/14/2021] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress is usually associated with prolonged intake of high-fat diet (HFD). However, little is known about the impact of maternal HFD on endogenous modulation of antioxidant-defence-enzyme-network, its link to adverse fetal growth and overall effects of Quercetin-3-o-rutinoside (QR) supplementation. Sprague-Dawley rats were initially assigned to normal diet (ND) or HFD for 8 weeks and mated. Post-conception, rats were further divided into four groups, of which two groups had diets supplemented with QR while others continued with their respective diets until delivery. Measurements include food and water consumption, physical parameters (body weight, body mass index (BMI) and fur appearance), oral glucose tolerance, lipid profiles, and placental/liver oxidative changes. We observed that water consumption was significantly increased in dams fed HFD without marked differences in food intake, body weight, BMI and glucose tolerance. Surprisingly, offspring of HFD-fed dams had reduced body weight marked by delayed fur appearance compared to the ND offspring. In dams, there were alterations in lipid profile. Lipid peroxidation was increased in the placenta and liver of gestational day (GD) 19 HFD-fed dams and their postnatal day (PND) 21 male offspring. There was evidence of HFD-induced nitrosative stress in dams and PND28 female offspring. Adaptive defence indicate decreased placenta and liver superoxide dismutase (SOD) levels as well as differential changes in total antioxidant capacity (TAC) and catalase (CAT) activity in HFD treated dams and their progenies. Overall, the results indicate that intrauterine metabolic alterations associated with maternal high-fat consumption may induce oxidative challenge in the offspring accompanied by mild developmental consequences, while QR supplementation has little or no beneficial effects.
Collapse
|
30
|
de Almeida-Faria J, Duque-Guimarães DE, Ong TP, Pantaleão LC, Carpenter AA, Loche E, Kusinski LC, Ashmore TJ, Antrobus R, Bushell M, Fernandez-Twinn DS, Ozanne SE. Maternal obesity during pregnancy leads to adipose tissue ER stress in mice via miR-126-mediated reduction in Lunapark. Diabetologia 2021; 64:890-902. [PMID: 33501603 PMCID: PMC7940301 DOI: 10.1007/s00125-020-05357-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/26/2020] [Indexed: 02/02/2023]
Abstract
AIMS/HYPOTHESIS Levels of the microRNA (miRNA) miR-126-3p are programmed cell-autonomously in visceral adipose tissue of adult offspring born to obese female C57BL/6J mice. The spectrum of miR-126-3p targets and thus the consequences of its dysregulation for adipocyte metabolism are unknown. Therefore, the aim of the current study was to identify novel targets of miR-126-3p in vitro and then establish the outcomes of their dysregulation on adipocyte metabolism in vivo using a well-established maternal obesity mouse model. METHODS miR-126-3p overexpression in 3T3-L1 pre-adipocytes followed by pulsed stable isotope labelling by amino acids in culture (pSILAC) was performed to identify novel targets of the miRNA. Well-established bioinformatics algorithms and luciferase assays were then employed to confirm those that were direct targets of miR-126-3p. Selected knockdown experiments were performed in vitro to define the consequences of target dysregulation. Quantitative real-time PCR, immunoblotting, histology, euglycaemic-hyperinsulinaemic clamps and glucose tolerance tests were performed to determine the phenotypic and functional outcomes of maternal programmed miR-126-3p levels in offspring adipose tissue. RESULTS The proteomic approach confirmed the identity of known targets of miR-126-3p (including IRS-1) and identified Lunapark, an endoplasmic reticulum (ER) protein, as a novel one. We confirmed by luciferase assay that Lunapark was a direct target of miR-126-3p. Overexpression of miR-126-3p in vitro led to a reduction in Lunapark protein levels and increased Perk (also known as Eif2ak3) mRNA levels and small interference-RNA mediated knockdown of Lunapark led to increased Xbp1, spliced Xbp1, Chop (also known as Ddit3) and Perk mRNA levels and an ER stress transcriptional response in 3T3-L1 pre-adipocytes. Consistent with the results found in vitro, increased miR-126-3p expression in adipose tissue from adult mouse offspring born to obese dams was accompanied by decreased Lunapark and IRS-1 protein levels and increased markers of ER stress. At the whole-body level the animals displayed glucose intolerance. CONCLUSIONS/INTERPRETATION Concurrently targeting IRS-1 and Lunapark, a nutritionally programmed increase in miR-126-3p causes adipose tissue insulin resistance and an ER stress response, both of which may contribute to impaired glucose tolerance. These findings provide a novel mechanism by which obesity during pregnancy leads to increased risk of type 2 diabetes in the offspring and therefore identify miR-126-3p as a potential therapeutic target.
Collapse
Affiliation(s)
- Juliana de Almeida-Faria
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, State University of Campinas, São Paulo, Brazil.
| | - Daniella E Duque-Guimarães
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Thomas P Ong
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Food Research Center, São Paulo, Brazil
| | - Lucas C Pantaleão
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Asha A Carpenter
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Elena Loche
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Laura C Kusinski
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Thomas J Ashmore
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge, UK
| | - Martin Bushell
- Cancer Research UK (CRUK), Beatson Institute, Glasgow, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
31
|
Abstract
The worldwide rates of obesity have increased significantly in recent decades. In the United States, more than 50% of pregnant women are overweight or obese. Obese gravid women are more prone to adverse pregnancy outcomes, including gestational diabetes, hypertensive disorders, and cardiovascular diseases. The adverse outcomes extend beyond the pregnant obese woman; offspring of obese women are themselves at increased risk of prematurity, fetal death, injury during birth, and transient respiratory problems and metabolic effects (ie, neonatal hypoglycemia). Furthermore, maternal obesity can predispose their offspring to long-term health problems, potentially generating an intergenerational cycle of obesity and insulin resistance.
Collapse
Affiliation(s)
- Carmen Paredes
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI.,Department of Obstetrics and Gynecology, Detroit Medical Center, Detroit, MI
| | - Richard C Hsu
- Wayne State University School of Medicine, Detroit, MI
| | - Anna Tong
- Wayne State University School of Medicine, Detroit, MI
| | - Jeffery R Johnson
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI.,Department of Obstetrics and Gynecology, Detroit Medical Center, Detroit, MI
| |
Collapse
|
32
|
Cattane N, Räikkönen K, Anniverno R, Mencacci C, Riva MA, Pariante CM, Cattaneo A. Depression, obesity and their comorbidity during pregnancy: effects on the offspring's mental and physical health. Mol Psychiatry 2021; 26:462-481. [PMID: 32632208 PMCID: PMC7850968 DOI: 10.1038/s41380-020-0813-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/20/2020] [Accepted: 06/05/2020] [Indexed: 02/08/2023]
Abstract
Depression and obesity represent two of the most common complications during pregnancy and are associated with severe health risks for both the mother and the child. Although several studies have analysed the individual effects of depression or obesity on the mothers and their children, the effects associated with the co-occurrence of both disorders have so far been poorly investigated. The relationship between depression and obesity is very complex and it is still unclear whether maternal depression leads to obesity or vice versa. It is well known that the intrauterine environment plays an important role in mediating the effects of both depression and obesity in the mother on the fetal programming, increasing the child's risk to develop negative outcomes.
Collapse
Affiliation(s)
- Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Roberta Anniverno
- Department of Neuroscience, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Claudio Mencacci
- Department of Neuroscience, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
33
|
Desoye G, Herrera E. Adipose tissue development and lipid metabolism in the human fetus: The 2020 perspective focusing on maternal diabetes and obesity. Prog Lipid Res 2020; 81:101082. [PMID: 33383022 DOI: 10.1016/j.plipres.2020.101082] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
During development, the human fetus accrues the highest proportion of fat of all mammals. Precursors of fat lobules can be found at week 14 of pregnancy. Thereafter, they expand, filling with triacylglycerols during pregnancy. The resultant mature lipid-filled adipocytes emerge from a developmental programme of embryonic stem cells, which is regulated differently than adult adipogenesis. Fetal triacylglycerol synthesis uses glycerol and fatty acids derived predominantly from glycolysis and lipogenesis in liver and adipocytes. The fatty acid composition of fetal adipose tissue at the end of pregnancy shows a preponderance of palmitic acid, and differs from the mother. Maternal diabetes mellitus does not influence this fatty acid profile. Glucose oxidation is the main source of energy for the fetus, but mitochondrial fatty acid oxidation also contributes. Indirect evidence suggests the presence of lipoprotein lipase in fetal adipose tissue. Its activity may be increased under hyperinsulinemic conditions as in maternal diabetes mellitus and obesity, thereby contributing to increased triacylglycerol deposition found in the newborns of such pregnancies. Fetal lipolysis is low. Changes in the expression of genes controlling metabolism in fetal adipose tissue appear to contribute actively to the increased neonatal fat mass found in diabetes and obesity. Many of these processes are under endocrine regulation, principally by insulin, and show sex-differences. Novel fatty acid derived signals such as oxylipins are present in cord blood with as yet undiscovered function. Despite many decades of research on fetal lipid deposition and metabolism, many key questions await answers.
Collapse
Affiliation(s)
- G Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria.
| | - E Herrera
- Faculties of Pharmacy and Medicine, University CEU San Pablo, Madrid, Spain.
| |
Collapse
|
34
|
Ardıç C, Çolak S, Uzun K, Salı G, Aydemir T, Telatar G. Maternal Gestational Diabetes and Early Childhood Obesity: A Retrospective Cohort Study. Child Obes 2020; 16:579-585. [PMID: 33146559 DOI: 10.1089/chi.2020.0183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Recently, childhood obesity has become one of the most serious public health problems in the world. Gestational diabetes mellitus (GDM) is considered a risk factor for childhood overweight and obesity. The study aimed at investigating the relationship between maternal GDM and childhood obesity in children aged from 1 to 3 years. Methods: In this retrospective cohort study, 237 GDM and 296 non-GDM mothers and their offspring who were followed up by Family Medicine Clinics in Rize province of Turkey were assessed. World Health Organization (WHO) criteria were used for the diagnosis of maternal GDM. Crude and adjusted logistic regression models were calculated for the association of gestational diabetes and childhood overweight/obesity. Gender and age-specific percentile tables were used for the categorization of BMI. Results: Statistical analysis carried out with adjustment for potential confounders (mother's age, educational status, smoking status, BMI, gestational weight gain, children's gender, and gestational birth weight) provided results with an odds ratio of 2.99; 95% CI 1.14-7.94 and 7.77; 95% CI 1.92-31.37 for the impact of gestational diabetes on childhood overweight and obesity at 2 and 3 years of age, respectively. Conclusions: This study found evidence for maternal GDM to cause the risk of early childhood obesity. Therefore, proper intervention strategies are required for this high-risk population.
Collapse
Affiliation(s)
- Cüneyt Ardıç
- Department of Family Medicine, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sabri Çolak
- Department of Obstetrics and Gynecology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerem Uzun
- Department of Family Medicine, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Görsel Salı
- Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Teslime Aydemir
- Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Gökhan Telatar
- Department of Public Health, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
35
|
Venkatratnam A, Douillet C, Topping BC, Shi Q, Addo KA, Ideraabdullah FY, Fry RC, Styblo M. Sex-dependent effects of preconception exposure to arsenite on gene transcription in parental germ cells and on transcriptomic profiles and diabetic phenotype of offspring. Arch Toxicol 2020; 95:473-488. [PMID: 33145626 DOI: 10.1007/s00204-020-02941-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/20/2020] [Indexed: 12/25/2022]
Abstract
Chronic exposure to inorganic arsenic (iAs) has been linked to diabetic phenotypes in both humans and mice. However, diabetogenic effects of iAs exposure during specific developmental windows have never been systematically studied. We have previously shown that in mice, combined preconception and in utero exposures to iAs resulted in impaired glucose homeostasis in male offspring. The goal of the present study was to determine if preconception exposure alone can contribute to this outcome. We have examined metabolic phenotypes in male and female offspring from dams and sires that were exposed to iAs in drinking water (0 or 200 μg As/L) for 10 weeks prior to mating. The effects of iAs exposure on gene expression profiles in parental germ cells, and pancreatic islets and livers from offspring were assessed using RNA sequencing. We found that iAs exposure significantly altered transcript levels of genes, including diabetes-related genes, in the sperm of sires. Notably, some of the same gene transcripts and the associated pathways were also altered in the liver of the offspring. The exposure had a more subtle effect on gene expression in maternal oocytes and in pancreatic islets of the offspring. In female offspring, the preconception exposure was associated with increased adiposity, but lower blood glucose after fasting and after glucose challenge. HOMA-IR, the indicator of insulin resistance, was also lower. In contrast, the preconception exposure had no effects on blood glucose measures in male offspring. However, males from parents exposed to iAs had higher plasma insulin after glucose challenge and higher insulinogenic index than control offspring, indicating a greater requirement for insulin to maintain glucose homeostasis. Our results suggest that preconception exposure may contribute to the development of diabetic phenotype in male offspring, possibly mediated through germ cell-associated inheritance. Future research can investigate role of epigenetics in this phenomenon. The paradoxical outcomes in female offspring, suggesting a protective effect of the preconception exposure, warrant further investigation.
Collapse
Affiliation(s)
- Abhishek Venkatratnam
- Department of Nutrition, CB# 7461, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7461, USA
- Department of Environmental Sciences and Engineering, CB#7431, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7431, USA
| | - Christelle Douillet
- Department of Nutrition, CB# 7461, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7461, USA
| | - Brent C Topping
- Department of Environmental Sciences and Engineering, CB#7431, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7431, USA
| | - Qing Shi
- Department of Nutrition, CB# 7461, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7461, USA
| | - Kezia A Addo
- Department of Environmental Sciences and Engineering, CB#7431, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7431, USA
| | - Folami Y Ideraabdullah
- Department of Nutrition, CB# 7461, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7461, USA
- Department of Genetics, CB#7264, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7264, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, CB#7431, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7431, USA.
| | - Miroslav Styblo
- Department of Nutrition, CB# 7461, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7461, USA.
| |
Collapse
|
36
|
Curci SG, Hernández JC, Luecken LJ, Perez M. Multilevel prenatal socioeconomic determinants of Mexican American children's weight: Mediation by breastfeeding. Health Psychol 2020; 39:997-1006. [PMID: 32969698 PMCID: PMC7919009 DOI: 10.1037/hea0001028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: Mexican American (MA) children are more likely to grow up in poverty than their non-Hispanic/Latinx white peers and are at an elevated risk for early onset obesity. The current study evaluated the effects of prenatal family- and neighborhood-level disadvantage on children's weight and weight gain from 12 months through 4.5 years of age. Maternal breastfeeding duration was evaluated as a potential mechanism underlying the relation between multilevel disadvantage and weight. Methods: Data was collected from 322 low-income, MA mother-child dyads. Women reported the degree of family socioeconomic disadvantage and breastfeeding status. Neighborhood disadvantage was evaluated with census-level metrics. Children's weight and height were measured at laboratory visits. Results: Greater prenatal neighborhood disadvantage predicted higher child Body Mass Index (BMI) at 12 months, over and above family-level disadvantage; this effect remained stable through 4.5 years. Breastfeeding duration partially mediated the effect of neighborhood disadvantage on child BMI. Breastfeeding duration predicted child BMI at all timepoints. Conclusions: Maternal prenatal residence in a neighborhood with high concentrated disadvantage may place low-income, MA children at increased risk of elevated weight status during the first few years of life. Breastfeeding duration emerged as potentially modifiable pathway through which the prenatal neighborhood impacts children's early life weight. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
|
37
|
Maternal high-fat diet induces long-term obesity with sex-dependent metabolic programming of adipocyte differentiation, hypertrophy and dysfunction in the offspring. Clin Sci (Lond) 2020; 134:921-939. [PMID: 32239178 DOI: 10.1042/cs20191229] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/19/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
Maternal obesity determines obesity and metabolic diseases in the offspring. The white adipose tissue (WAT) orchestrates metabolic pathways, and its dysfunction contributes to metabolic disorders in a sex-dependent manner. Here, we tested if sex differences influence the molecular mechanisms of metabolic programming of WAT in offspring of obese dams. To this end, maternal obesity was induced with high-fat diet (HFD) and the offspring were studied at an early phase [postnatal day 21 (P21)], a late phase (P70) and finally P120. In the early phase we found a sex-independent increase in WAT in offspring of obese dams using magnetic resonance imaging (MRI), which was more pronounced in females than males. While the adipocyte size increased in both sexes, the distribution of WAT differed in males and females. As mechanistic hints, we identified an inflammatory response in females and a senescence-associated reduction in the preadipocyte factor DLK in males. In the late phase, the obese body composition persisted in both sexes, with a partial reversal in females. Moreover, female offspring recovered completely from both the adipocyte hypertrophy and the inflammatory response. These findings were linked to a dysregulation of lipolytic, adipogenic and stemness-related markers as well as AMPKα and Akt signaling. Finally, the sex-dependent metabolic programming persisted with sex-specific differences in adipocyte size until P120. In conclusion, we do not only provide new insights into the molecular mechanisms of sex-dependent metabolic programming of WAT dysfunction, but also highlight the sex-dependent development of low- and high-grade pathogenic obesity.
Collapse
|
38
|
Campodonico-Burnett W, Hetrick B, Wesolowski SR, Schenk S, Takahashi DL, Dean TA, Sullivan EL, Kievit P, Gannon M, Aagaard K, Friedman JE, McCurdy CE. Maternal Obesity and Western-Style Diet Impair Fetal and Juvenile Offspring Skeletal Muscle Insulin-Stimulated Glucose Transport in Nonhuman Primates. Diabetes 2020; 69:1389-1400. [PMID: 32354857 PMCID: PMC7306120 DOI: 10.2337/db19-1218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/28/2020] [Indexed: 12/12/2022]
Abstract
Infants born to mothers with obesity have a greater risk for childhood obesity and metabolic diseases; however, the underlying biological mechanisms remain poorly understood. We used a Japanese macaque model to investigate whether maternal obesity combined with a Western-style diet (WSD) impairs offspring muscle insulin action. Adult females were fed a control or WSD prior to and during pregnancy through lactation, and offspring subsequently weaned to a control or WSD. Muscle glucose uptake and signaling were measured ex vivo in fetal (n = 5-8/group) and juvenile (n = 8/group) offspring. In vivo signaling was evaluated after an insulin bolus just prior to weaning (n = 4-5/group). Maternal WSD reduced insulin-stimulated glucose uptake and impaired insulin signaling at the level of Akt phosphorylation in fetal muscle. In juvenile offspring, insulin-stimulated glucose uptake was similarly reduced by both maternal and postweaning WSD and corresponded to modest reductions in insulin-stimulated Akt phosphorylation relative to controls. We conclude that maternal WSD leads to a persistent decrease in offspring muscle insulin-stimulated glucose uptake even in the absence of increased offspring adiposity or markers of systemic insulin resistance. Switching offspring to a healthy diet did not reverse the effects of maternal WSD on muscle insulin action, suggesting earlier interventions may be warranted.
Collapse
Affiliation(s)
- William Campodonico-Burnett
- Department of Human Physiology, University of Oregon, Eugene, OR
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, OR
| | | | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health Science University, Beaverton, OR
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health Science University, Beaverton, OR
| | - Elinor L Sullivan
- Department of Human Physiology, University of Oregon, Eugene, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health Science University, Beaverton, OR
- Department of Psychiatry, Oregon Health Science University, Portland, OR
- Department of Behavioral Sciences, Oregon Health Science University, Portland, OR
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health Science University, Beaverton, OR
| | - Maureen Gannon
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Kjersti Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR
| |
Collapse
|
39
|
Chowdhury R, Taneja S, Dhabhai N, Mazumder S, Upadhyay RP, Sharma S, Tupaki-Sreepurna A, Dewan R, Mittal P, Chellani H, Bahl R, Bhan MK, Bhandari N. Burden of preconception morbidity in women of reproductive age from an urban setting in North India. PLoS One 2020; 15:e0234768. [PMID: 32555694 PMCID: PMC7302496 DOI: 10.1371/journal.pone.0234768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 06/01/2020] [Indexed: 12/26/2022] Open
Abstract
Background There is a growing interest in the life course approach for the prevention, early detection and subsequent management of morbidity in women of reproductive age to ensure optimal health and nutrition when they enter pregnancy. Reliable estimates of such morbidities are lacking. We report the prevalence of health or nutrition-related morbidities, specifically, anemia, undernutrition, overweight and obesity, sexually transmitted infections (STIs) or reproductive tract infections (RTIs), diabetes or prediabetes, hypothyroidism, hypertension, and depressive symptoms, during the preconception period among women aged 18 to 30 years. Methods A cross-sectional study was conducted among 2000 nonpregnant married women aged 18 to 30 years with no or one child who wished to have more children in two low- to middle-income urban neighborhoods in Delhi, India, in the context of a randomized controlled trial. STIs and RTIs were measured by symptoms and signs, blood pressure by a digital device, height by stadiometer and weight by a digital weighing scale. A blood specimen was taken to screen for anemia, diabetes, thyroid disorders and syphilis. Maternal depressive symptoms were assessed using the Patient Health Questionnaire-9 (PHQ-9). Multivariable logistic regression analysis was performed to identify sociodemographic factors associated with individual morbidity. Results Overall, 58.7% of women were anemic; 16.5%, undernourished; 26%, overweight or obese; 13.2%, hypothyroid; and 10.5% with both symptoms and signs of STIs/RTIs. There was an increased risk of RTI/STI symptoms and signs in undernourished women and an increased risk of diabetes or prediabetes in overweight or obese women. An increased risk of undernutrition was also observed in women from lower categories of wealth quintiles. A decreased risk of moderate to severe anemia was seen in overweight women and those who completed at least secondary education. Conclusions Our findings show a high burden of undernutrition, anemia, RTIs, hypothyroidism and prediabetes among women in the study. This information will aid policymakers in planning special programs for women of reproductive age.
Collapse
Affiliation(s)
- Ranadip Chowdhury
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Sunita Taneja
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Neeta Dhabhai
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Sarmila Mazumder
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Ravi Prakash Upadhyay
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Sitanshi Sharma
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | - Rupali Dewan
- Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Pratima Mittal
- Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Harish Chellani
- Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Rajiv Bahl
- Department of Maternal, Newborn, Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| | - Maharaj Kishan Bhan
- Knowledge Integration and Translational Platform (KnIT), Biotechnology Industry Research Assistance Council (BIRAC), Department of Biotechnology, Government of India, New Delhi, India
| | - Nita Bhandari
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
- * E-mail:
| |
Collapse
|
40
|
Fetal macrosomia in a Hispanic/Latinx predominant cohort and altered expressions of genes related to placental lipid transport and metabolism. Int J Obes (Lond) 2020; 44:1743-1752. [PMID: 32494035 PMCID: PMC7387181 DOI: 10.1038/s41366-020-0610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/17/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Fetal overgrowth, termed fetal macrosomia when birth weight is greater than 4000 grams, is the major concern in the treatment of gestational diabetes mellitus (GDM). However, to date, the underlying mechanisms of fetal macrosomia have not been understood completely. Placental lipid metabolism is emerging as a critical player in fetal growth. In this study, we hypothesized that fatty acid transport and metabolism in the placental tissue was impaired in GDM women, dependent on fetal sex. To test this hypothesis, we analyzed the incidence of GDM, fetal macrosomia, and obesity in a large cohort consisting of 17995 pregnant subjects and majority of subjects being Hispanic/Latinx, and investigated expression of genes related to lipid transport and metabolism in placenta from obese women with or without GDM, and with or without fetal macrosomia. The main findings include: 1) There is a higher incidence of GDM and obesity in Hispanic subjects compared to non-Hispanic subjects, but not fetal macrosomia; 2) Expressions of most of genes related to placental lipid transport and metabolism are not altered by the presence of GDM, fetal macrosomia, or fetal sex; 3) Expression of FABP4 is increased in obese women with GDM and fetal macrosomia, and this occurred in male placentas; 4) Expression of LPL is decreased in obese women with GDM despite fetal macrosomia, and this occurred in male placentas; 5) Expression of ANGPTL3 is decreased in obese women with GDM and fetal macrosomia, but is not altered when fetal sex is included in the analysis. This study indicates that there is race disparity in GDM with higher incidence of GDM in obese Hispanic women, although fetal macrosomia disparity is not present. Moreover, altered placental lipid transport may contribute to fetal overgrowth in obese women with GDM.
Collapse
|
41
|
Puttabyatappa M, Sargis RM, Padmanabhan V. Developmental programming of insulin resistance: are androgens the culprits? J Endocrinol 2020; 245:R23-R48. [PMID: 32240982 PMCID: PMC7219571 DOI: 10.1530/joe-20-0044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
Insulin resistance is a common feature of many metabolic disorders. The dramatic rise in the incidence of insulin resistance over the past decade has enhanced focus on its developmental origins. Since various developmental insults ranging from maternal disease, stress, over/undernutrition, and exposure to environmental chemicals can all program the development of insulin resistance, common mechanisms may be involved. This review discusses the possibility that increases in maternal androgens associated with these various insults are key mediators in programming insulin resistance. Additionally, the intermediaries through which androgens misprogram tissue insulin sensitivity, such as changes in inflammatory, oxidative, and lipotoxic states, epigenetic, gut microbiome and insulin, as well as data gaps to be filled are also discussed.
Collapse
Affiliation(s)
| | - Robert M. Sargis
- Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | | |
Collapse
|
42
|
Daniels TE, Sadovnikoff AI, Ridout KK, Lesseur C, Marsit CJ, Tyrka AR. Associations of maternal diet and placenta leptin methylation. Mol Cell Endocrinol 2020; 505:110739. [PMID: 32004678 PMCID: PMC7185035 DOI: 10.1016/j.mce.2020.110739] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Maternal diet is an important factor in prenatal development that also has implications for disease risk later in life. The adipokine leptin is a key regulator of energy homeostasis and may be involved in the association between maternal nutrition, maternal obesity, and infant outcomes. DNA methylation of placenta genes may occur in response to exposures and may program subsequent infant development. This study examined maternal diet, placenta leptin gene DNA methylation, and neonatal growth in a sample of healthy neonates and their mothers. METHODS Mothers and their healthy neonates (N = 135) were recruited within 1-2 days following delivery at Women and Infants Hospital in Providence, RI. A structured interview was conducted to assess maternal dietary intake. Maternal pre-pregnancy weight, weight gain during pregnancy, maternal health, medications, and vitamin use were obtained from medical records. Bisulfite pyrosequencing was used to measure methylation of CpG sites in the promoter region of the placenta leptin gene and determine genotype of the leptin single nucleotide polymorphism (SNP) rs2167270, which is known to influence leptin methylation. Bivariate analyses and linear regression models were used to evaluate associations of demographics, diet, and mean leptin methylation. RESULTS Genotype was a significant predictor of placenta leptin DNA methylation (p < .01), and after controlling for this and other relevant maternal and infant covariates, lower levels of leptin methylation were significantly associated with greater intake of carbohydrates (p < .05), in particular added sugars (p < .05) and white/refined carbohydrates (p < .05). Total caloric intake was also associated with placenta leptin methylation (p < .05), however after controlling for relevant covariates, significance diminished to trend-level. There were no significant associations of placenta leptin methylation and intake of protein (p > .05) or fat (p > .05). CONCLUSION These findings underline the importance of intake of carbohydrate consumption for methylation of the placenta leptin gene. Because methylation reduces gene transcription, lower methylation may indicate a placenta response to high caloric intake and carbohydrate food that would result in higher levels of this hormone during fetal development. Further investigation of the developmental ramifications of epigenetic changes to placenta leptin methylation should be pursued.
Collapse
Affiliation(s)
- Teresa E Daniels
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, USA.
| | - Alexander I Sadovnikoff
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA
| | - Kathryn K Ridout
- Department of Psychiatry and Family Medicine, Kaiser Permanente, San Jose, CA, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, USA
| | - Audrey R Tyrka
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, USA
| |
Collapse
|
43
|
Nicholas LM, Ozanne SE. Early life programming in mice by maternal overnutrition: mechanistic insights and interventional approaches. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180116. [PMID: 30966886 DOI: 10.1098/rstb.2018.0116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models have been indispensable in elucidating the potential causative mechanisms underlying the effects of maternal diet on offspring health. Of these, the mouse has been widely used to model maternal overnutrition and/or maternal obesity and to study its effects across one or more generations. This review discusses recent findings from mouse models, which resemble the human situation, i.e. overnutrition/obesity across pregnancy and lactation. It also highlights the importance of embryo transfer models in identifying critical developmental period(s) during which specific metabolic changes are programmed in the offspring. The mouse is also an excellent tool for maternal intervention studies aimed at elucidating the longer-term effects on the offspring and for defining possible maternal factors underling the programming of metabolic adversity in offspring. While knowledge of the mouse genome and the molecular tools available have allowed great progress to be made in the field, it is clear that we need to define if the effects on the offspring are mediated by maternal obesity per se or if specific components of the maternal metabolic environment are more important. We can then begin to identify at-risk offspring and to design more effective interventions for the mother and/or her child. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Lisa M Nicholas
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| | - Susan E Ozanne
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| |
Collapse
|
44
|
Insulin Resistance in Pregnancy: Implications for Mother and Offspring. CONTEMPORARY ENDOCRINOLOGY 2020. [DOI: 10.1007/978-3-030-25057-7_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Cheng C, Wu X, Zhang X, Zhang X, Peng J. Obesity of Sows at Late Pregnancy Aggravates Metabolic Disorder of Perinatal Sows and Affects Performance and Intestinal Health of Piglets. Animals (Basel) 2019; 10:ani10010049. [PMID: 31881697 PMCID: PMC7023453 DOI: 10.3390/ani10010049] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Our novel findings suggest that excessive backfat thickness of sows at days 109 of gestation exacerbates the metabolic disorder of perinatal sows, reduces the number and litter weight of piglets born alive, and adversely affects the intestinal health of sows and their offspring piglets. Moreover, the current study also provides an important theoretical reference for strengthening the control of body condition in sows during reproductive cycle. Abstract This study explored the effect of obesity of sows in late pregnancy on metabolic status of perinatal sows and performance, intestinal health, and immune system of offspring piglets. Sixty multiparous Landrance × Large White sows were selected in this study. Sows were divided into two groups according to backfat thickness (normal backfat thickness group, =17 mm; excessive backfat thickness group, ≥21 mm) at days 109 of gestation. The excessive backfat thickness of sows during late pregnancy decreased the total number and litter weight of piglets born alive. Compared with normal backfat thickness sows, the excessive backfat thickness sows had increased levels of plasma glucose, IL-6, and TNF-α and homeostasis model assessment insulin resistance values. The excessive backfat thickness also reduced total superoxide dismutase but increased thiobarbituric acid reactive substances in plasma of perinatal sows. Additionally, the fecal levels of TNF-α were increased but those of IL-10 were decreased in piglets from excessive backfat thickness sow. These findings indicate that the obesity of sows during late pregnancy aggravates the metabolic disorder of perinatal sows, reduces the number of piglets born alive, and adversely affects the intestinal health of sows and their offspring piglets.
Collapse
Affiliation(s)
- Chuanshang Cheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.C.); (X.W.); (X.Z.)
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, WENS Research Institute (Technology Center), Yunfu 527300, China
| | - Xiaoyu Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.C.); (X.W.); (X.Z.)
| | - Xiaofeng Zhang
- College of Life Sciences, Zhaoqing University, Zhaoqing 526061, China;
| | - Xiu Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.C.); (X.W.); (X.Z.)
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.C.); (X.W.); (X.Z.)
- The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
46
|
Kadakia R, Zheng Y, Zhang Z, Zhang W, Josefson JL, Hou L. Association of cord blood methylation with neonatal leptin: An epigenome wide association study. PLoS One 2019; 14:e0226555. [PMID: 31851703 PMCID: PMC6919608 DOI: 10.1371/journal.pone.0226555] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022] Open
Abstract
Background Neonatal adiposity is a risk factor for childhood obesity. Investigating contributors to neonatal adiposity is important for understanding early life obesity risk. Epigenetic changes of metabolic genes in cord blood may contribute to excessive neonatal adiposity and subsequent childhood obesity. This study aims to evaluate the association of cord blood DNA methylation patterns with anthropometric measures and cord blood leptin, a biomarker of neonatal adiposity. Methods A cross-sectional study was performed on a multiethnic cohort of 114 full term neonates born to mothers without gestational diabetes at a university hospital. Cord blood was assayed for leptin and for epigenome-wide DNA methylation profiles via the Illumina 450K platform. Neonatal body composition was measured by air displacement plethysmography. Multivariable linear regression was used to analyze associations between individual CpG sites as well as differentially methylated regions in cord blood DNA with measures of newborn adiposity including anthropometrics (birth weight, fat mass and percent body fat) and cord blood leptin. False discovery rate was estimated to account for multiple comparisons. Results 247 CpG sites as well as 18 differentially methylated gene regions were associated with cord blood leptin but no epigenetic changes were associated with birth weight, fat mass or percent body fat. Genes of interest identified in this study are DNAJA4, TFR2, SMAD3, PLAG1, FGF1, and HNF4A. Conclusion Epigenetic changes in cord blood DNA are associated with cord blood leptin levels, a measure of neonatal adiposity.
Collapse
Affiliation(s)
- Rachel Kadakia
- Division of Endocrinology, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| | - Yinan Zheng
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Zhou Zhang
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Wei Zhang
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Jami L. Josefson
- Division of Endocrinology, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Lifang Hou
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
47
|
Yuan X, Gao Y, Zhang M, Long W, Liu J, Wang H, Yu B, Xu J. Association of maternal D-dimer level in late pregnancy with birth outcomes in a Chinese cohort. Clin Chim Acta 2019; 501:258-263. [PMID: 31730819 DOI: 10.1016/j.cca.2019.10.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the association of D-dimer level during late pregnancy with birth outcomes in a Chinese population. METHODS A retrospective observational cohort study of 11,570 pregnant women who delivered in a single central hospital was conducted. Maternal plasma D-dimer levels at hospital admission and pregnancy outcomes were abstracted and analyzed from laboratory information system and hospital records, respectively. RESULTS Maternal plasma D-dimer levels were associated with higher fetal growth indicators for the highest vs. lowest quartile (Q) of D-dimer (mean birth weight: 145.79 g, mean birth length: 0.11 cm, mean gestational age: 0.30 week). Increase in D-dimer quartiles were associated with an decreased risk for small for gestational age (SGA), low birth weight (LBW) and preterm birth (PTB) neonates, and with an increased risk for large for gestational age (LGA), and macrosomia infants (SGA: OR = 0.52, 95% CI: 0.43, 0.64; LBW: OR = 0.58, 95% CI: 0.38, 0.86; PTB: OR = 0.44, 95% CI: 0.35, 0.55; LGA: OR = 2.37, 95% CI: 2.01, 2.78; macrosomia: OR = 2.59, 95% CI, 2.06, 3.24; for Q4 vs. Q1). CONCLUSION Maternal plasma D-dimer levels during late pregnancy were associated with birth outcomes and had risk evaluation value for these outcomes.
Collapse
Affiliation(s)
- Xiaosong Yuan
- Department of Medical Genetics, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China
| | - Yanfang Gao
- Department of Laboratory Medicine, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China.
| | - Ming Zhang
- Department of Laboratory Medicine, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China
| | - Wei Long
- Department of Medical Genetics, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China
| | - Jianbing Liu
- Department of Medical Genetics, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China
| | - Huiyan Wang
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China
| | - Bin Yu
- Department of Medical Genetics, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China.
| | - Jun Xu
- Department of Medical Genetics, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, No. 16 Ding Xiang Road, Changzhou, Jiangsu Province, China.
| |
Collapse
|
48
|
Silva LBAR, Pinheiro-Castro N, Novaes GM, Pascoal GDFL, Ong TP. Bioactive food compounds, epigenetics and chronic disease prevention: Focus on early-life interventions with polyphenols. Food Res Int 2019; 125:108646. [PMID: 31554120 DOI: 10.1016/j.foodres.2019.108646] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
|
49
|
Gao H, Li P, Liu L, Yang K, Xiao B, Zhou G, Tian Z, Luo C, Xia T, Dong L, Zhao Q, Wang A, Zhang S. Perigestational low-dose BDE-47 exposure alters maternal serum metabolome and results in sex-specific weight gain in adult offspring. CHEMOSPHERE 2019; 233:174-182. [PMID: 31173955 DOI: 10.1016/j.chemosphere.2019.05.277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/25/2019] [Accepted: 05/29/2019] [Indexed: 06/09/2023]
Abstract
Emerging evidence suggests environmental contaminant exposures during critical windows of development may contribute to the increasing prevalence of obesity. It has been shown that early life polybrominated diphenyl ethers exposures have critical impacts on child weight trajectories, however, little is known about their maternal mechanisms responsible for offspring obesity development. In this study, we investigated the effects of perigestational low-dose 2, 2', 4, 4'-tetrabromodiphenyl ether (BDE-47) exposure on maternal metabolome, and its possible link to adult offspring bodyweight changes. Female Sprague-Dawley rats were exposed to daily doses of 0.1, or 1 mg/kg BDE-47 from 10 days prior to conception until offspring were weaned on postnatal day 21, and then a gas chromatography-mass spectrometry based metabolomics analysis was used to uncover the global metabolic response in dams. The pups continued to grow into adulthood for measurements of bodyweight. Perigestational BDE-47 exposure caused increased adult bodyweight in male but not in female offspring and dams. Metabolomics revealed significant changes in maternal serum metabolites that clearly distinguish BDE-47 from control rats. These differentially expressed metabolites were primarily implicated in amino acid, lipid, carbohydrate, and energy metabolisms, which was confirmed by pathway analysis. Importantly, most of these identified metabolites were decreased, a state similar to maternal malnutrition that can predispose adult male offspring to weight increase and adiposity in a postnatal environment with abundant calories. Collectively, our data suggest that perigestational exposure to low-dose BDE-47 produces altered maternal serum metabolome, which may be an additional contributing factor to weight gain in adult male offspring.
Collapse
Affiliation(s)
- Hui Gao
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pei Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Luming Liu
- Department of Medical Records and Statistics, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Kaichao Yang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Boya Xiao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Guoyu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Zhiyuan Tian
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chen Luo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Tao Xia
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Lixin Dong
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qian Zhao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Aiguo Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Shun Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
50
|
Kadakia R, Talbot O, Kuang A, Bain JR, Muehlbauer MJ, Stevens RD, Ilkayeva OR, Lowe LP, Metzger BE, Newgard CB, Scholtens DM, Lowe WL. Cord Blood Metabolomics: Association With Newborn Anthropometrics and C-Peptide Across Ancestries. J Clin Endocrinol Metab 2019; 104:4459-4472. [PMID: 31498869 PMCID: PMC6735762 DOI: 10.1210/jc.2019-00238] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022]
Abstract
CONTEXT Newborn adiposity is associated with childhood obesity. Cord blood metabolomics is one approach that can be used to understand early-life contributors to adiposity and insulin resistance. OBJECTIVE To determine the association of cord blood metabolites with newborn adiposity and hyperinsulinemia in a multiethnic cohort of newborns. DESIGN Cross-sectional, observational study. SETTING Hyperglycemia and Adverse Pregnancy Outcome study. PARTICIPANTS One thousand six hundred multiethnic mother-newborn pairs. MAIN OUTCOME MEASURE Cord blood C-peptide, birthweight, and newborn sum of skinfolds. RESULTS Meta-analyses across four ancestry groups (Afro-Caribbean, Northern European, Thai, and Mexican American) demonstrated significant associations of cord blood metabolites with cord blood C-peptide, birthweight, and newborn sum of skinfolds. Several metabolites, including branched-chain amino acids (BCAAs), medium- and long-chain acylcarnitines, nonesterified fatty acids, and triglycerides were negatively associated with cord C-peptide but positively associated with birthweight and/or sum of skinfolds. 1,5-Anhydroglucitol, an inverse marker of recent maternal glycemia, was significantly inversely associated with birthweight and sum of skinfolds. Network analyses revealed groups of interrelated amino acid, acylcarnitine, and fatty acid metabolites associated with all three newborn outcomes. CONCLUSIONS Cord blood metabolites are associated with newborn size and cord blood C-peptide levels after adjustment for maternal body mass index and glucose during pregnancy. Negative associations of metabolites with C-peptide at birth were observed. 1,5-Anhydroglucitol appears to be a marker of adiposity in newborns. BCAAs were individually associated with birthweight and demonstrated possible associations with newborn adiposity in network analyses.
Collapse
Affiliation(s)
- Rachel Kadakia
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
- Correspondence and Reprint Requests: William L. Lowe, Jr., MD, Feinberg School of Medicine, Northwestern University, Rubloff Building, 12th Floor, 420 East Superior Street, Chicago, Ilinois 60611.
| | - Octavious Talbot
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Alan Kuang
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Robert D Stevens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Lynn P Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Boyd E Metzger
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | | | - William L Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|