1
|
Li S, Zhou L, Ren J, Liu J, Zhang Q, Xiao X. The role of cecal metabolites and DNA methylation in deciphering the effects of maternal genistein intake on white fat browning in adult female offspring. Food Funct 2025; 16:3090-3100. [PMID: 40146209 DOI: 10.1039/d4fo04761c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Maternal diets during pregnancy and lactation are critical determinants that regulate the metabolic homeostasis in offspring. Our previous research demonstrated that maternal genistein (GEN) intervention ameliorated the dysregulation of glucolipid metabolism induced by intrauterine overnutrition in adult offspring, accompanied by changes in the composition of gut microbiota; however, the underlying mechanisms remain unclear. Here, we used a maternal overnutrition model induced by excess energy intake before and throughout pregnancy and lactation, with maternal GEN administered during the same period. The female offspring were raised on a standard chow diet until sacrificed at 24 weeks. The mRNA levels of browning markers were quantified in inguinal subcutaneous adipose tissues, followed by methylation analysis via the MassArray method. Cecal contents were collected for untargeted metabolomic analysis and a target quantitative analysis of methionine cycle metabolites. Spearman correlation analyses were used to assess whether cecal metabolites are involved in the methylation of browning-related genes and influence their expression. The results showed that maternal GEN supplementation reversed the downregulation of browning markers caused by perinatal high-fat diets in adult female offspring, consistent with a reduction in their methylation levels. Subsequently, we also found that maternal GEN consumption altered cecal metabolite profiles in offspring, promoting the production of bile acids, potent regulators of glucolipid metabolism, and reducing metabolites involved in the methionine cycle, key methyl donors for the methylation process. Furthermore, the abundances of these metabolites were significantly correlated with the methylation and expression levels of browning markers. Overall, this discovery suggested that maternal GEN intake decreased the methylation level of browning markers and induced browning in white adipose tissue of offspring, which correlated with alterations in cecal metabolites. We provide a novel theoretical basis for GEN as a promising nutritional supplement to break the vicious cycle of maternal metabolic disturbances being transmitted to offspring.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
2
|
Ding L, Liu J, Zhou L, Zhang Q, Liu J, Xiao X. Maternal high-fat diet alters the transcriptional rhythm in white adipose tissue of adult offspring. J Nutr Biochem 2025; 138:109843. [PMID: 39826765 DOI: 10.1016/j.jnutbio.2025.109843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
A maternal high-fat diet (HFD) deteriorates the long-term metabolic health of offspring. Circadian rhythms are crucial for regulating metabolism. However, the impact of maternal HFD on the circadian clock in white adipose tissue (WAT) remains unexplored. This study aimed to identify transcriptional rhythmic alterations in inguinal WAT of adult male offspring induced by maternal HFD. To this end, female mice were fed an HFD and their male offspring were raised on a standard chow diet until 16 weeks of age. Transcriptome was performed and the data was analyzed using CircaCompare. The results showed that maternal HFD before and throughout pregnancy significantly altered the circadian rhythm of inguinal WAT while slightly modifying the WAT clock in adult male offspring. Specifically, maternal HFD contributed to gaining rhythmicity of Cry2, resulted in the elevated amplitude of Nr1d2, and led to increased midline estimating statistic of rhythm (MESOR) of Clock and Nr1d2. Furthermore, maternal HFD changed the rhythmic pattern of metabolic genes, such as Pparγ, Hacd2, and Acsl1, which are significantly enriched in metabolic regulation pathways. In conclusion, a maternal HFD before and throughout pregnancy altered the circadian rhythm of inguinal WAT in adult offspring. These alterations may play a significant role in disturbing metabolic homeostasis, potentially leading to metabolic dysfunction in adult male offspring.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liyuan Zhou
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Nakayama K, Kaneko N, Hishimura N, Yamaguchi T, Morikawa S, Yokota I, Kamasaki H, Nagasaki K, Hasegawa Y, Nakamura A. Adiposity rebound and body mass index in Japanese patients with congenital hypothyroidism. Clin Pediatr Endocrinol 2025; 34:121-130. [PMID: 40201374 PMCID: PMC11972873 DOI: 10.1297/cpe.2024-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/21/2025] [Indexed: 04/10/2025] Open
Abstract
The long-term prognosis of congenital hypothyroidism (CH) has become apparent since the introduction of newborn screening programs; however, the risk of obesity in patients with CH remains unclear. Early adiposity rebound (AR) is one of the predictors of obesity in adults. This study evaluated AR and the adolescent body mass index (BMI) in Japanese patients with CH. We longitudinally collected anthropometric measurements from 288 patients aged 1-10 yr and plotted their BMI curves to determine the age at onset of AR. We also evaluated the effects of thyroid function, presence of distal femoral epiphysis (DFE) ossification, and disease type on AR age and adolescent BMI. The mean AR ages were determined to be 5.5 ± 1.4 yr in boys and 5.9 ± 1.5 yr in girls. There were no significant differences in AR age or adolescent BMI according to thyroid-stimulating hormone or free T4 levels before treatment initiation or according to disease type. However, at the last visit, more than half of the boys without DFE ossification had higher BMI SD scores than those with DFE ossification. These findings raise the possibility that severe prolonged fetal hypothyroidism may have a lasting influence after birth despite early treatment initiation.
Collapse
Affiliation(s)
- Kanako Nakayama
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Naoya Kaneko
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nozomi Hishimura
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takeshi Yamaguchi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Shuntaro Morikawa
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Isao Yokota
- Department of Biostatistics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hotaka Kamasaki
- Department of Pediatrics, Sapporo Medical University, Sapporo, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | - Yukihiro Hasegawa
- Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Akie Nakamura
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Molecular Endocrinology, National Center for Child Health and Development Research Center, Tokyo, Japan
| |
Collapse
|
4
|
Li S, Zhou L, Ren J, Zhang Q, Xiao X. Maternal exercise programs placental miR-495-5p-mediated Snx7 expression and kynurenic acid metabolic pathway induced by prenatal high-fat diet: Based on miRNA-seq, transcriptomics, and metabolomics. J Nutr Biochem 2025; 137:109830. [PMID: 39647668 DOI: 10.1016/j.jnutbio.2024.109830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Poor intrauterine environments increase the prevalence of chronic metabolic diseases in offspring, whereas maternal exercise is an effective measure to break this vicious intergenerational cycle. Placenta is increasingly being studied to explore its role in maternal-fetal metabolic cross-talk. The association between placental miRNA and offspring development trajectories has been established, yet the specific role and mechanism thereof in maternal exercise-induced metabolic protection remain elusive. Here, C57BL/6 female mice were subjected to either a normal control or a high-fat diet (HFD), half of the HFD-fed dams were housed with voluntary wheel running for 3 weeks before and during gestation. At embryonic day 18.5, we sacrificed parturient mice and then conducted miRNA-seq, transcriptomic, and metabolomic profiling of the placenta. Our data revealed that maternal HFD resulted in significant alterations in both miRNA and gene expressions, as well as metabolic pathways of the placenta, whereas prenatal exercise negated these perturbations. The common differentially expressed transcripts among three groups were enriched in multiple critical pathways involving energy expenditure, signal transduction, and fetal development. Through integrated analysis of multiomics data, we speculated that maternal exercise reversed the suppression of miR-495-5p induced by HFD, thereby inhibiting miR-495-5p-targeted Snx7 and modulating kynurenic acid production. These datasets provided novel mechanistic insight into how maternal exercise positively affects the metabolic homeostasis of offspring. The discovered important miRNAs, mRNAs, and metabolites could be promising predictive and therapeutic targets for protecting offspring metabolic health.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
5
|
Zhang Y, Hu S, Han S, Liu C, Liang X, Li Y, Lin Z, Qin Y, Geng C, Liu Y, Cui L, Hu J, Zhang C, Wang Z, Liu X, Ma J, Chen ZJ, Zhao H. Transgenerational inheritance of diabetes susceptibility in male offspring with maternal androgen exposure. Cell Discov 2025; 11:14. [PMID: 39934105 DOI: 10.1038/s41421-025-00769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025] Open
Abstract
Androgen exposure (AE) poses a profound health threat to women, yet its transgenerational impacts on male descendants remain unclear. Here, employing a large-scale mother-child cohort, we show that maternal hyperandrogenism predisposes sons to β-cell dysfunction. Male offspring mice with prenatal AE exhibited hyperglycemia and glucose intolerance across three generations, which were further exacerbated by aging and a high-fat diet. Mechanistically, compromised insulin secretion underlies this transgenerational susceptibility to diabetes. Integrated analyses of methylome and transcriptome revealed differential DNA methylation of β-cell functional genes in AE-F1 sperm, which was transmitted to AE-F2 islets and further retained in AE-F2 sperm, leading to reduced expression of genes related to insulin secretion, including Pdx1, Irs1, Ptprn2, and Cacna1c. The methylation signatures in AE-F1 sperm were corroborated in diabetic humans and the blood of sons with maternal hyperandrogenism. Moreover, caloric restriction and metformin treatments normalized hyperglycemia in AE-F1 males and blocked their inheritance to offspring by restoring the aberrant sperm DNA methylations. Our findings highlight the transgenerational inheritance of impaired glucose homeostasis in male offspring from maternal AE via DNA methylation changes, providing methylation biomarkers and therapeutic strategies to safeguard future generations' metabolic health.
Collapse
Affiliation(s)
- Yuqing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China.
| | - Shourui Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Shan Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Congcong Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Xiaofan Liang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Yuxuan Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Zongxuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Yiming Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Chunxuan Geng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Yue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
| | - Linlin Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Jingmei Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Changming Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Zhao Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Xin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Jinlong Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, the Second Hospital, Shandong University, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education (Shandong University), Ministry of Education, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China.
| |
Collapse
|
6
|
Andrade DC, Gaetani B, Moura S, de Carvalho SN, Thole AA, Cortez E. Maternal obesity negatively impacts cardiac progenitor cell survival in heart adulthood offspring. Nutr Metab Cardiovasc Dis 2025:103903. [PMID: 39988509 DOI: 10.1016/j.numecd.2025.103903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND AND AIM Maternal obesity has been related to offspring predisposition to cardiometabolic disease development throughout life. Thus, this study aimed to analyze the impact of maternal obesity on cardiac progenitor cells and cardiometabolic disease of adult offspring. METHODS AND RESULTS The litter size reduction experimental model was used to induce obesity in female Swiss mice. Small Litter Dam (SLD-F1) and Normal Litter Dam (NLD-F1) were crossed with healthy male mice, and their offspring were followed up until 90 days old when they were euthanized. Adult offspring of obese dams (SLD-F2) had increased body mass, Lee Index and fat deposits. Heart analysis demonstrated cardiac hypertrophy, fibrosis, oxidative stress, increased cardiac mast cell number, decreased cellular proliferation, increased proinflammatory cytokines, and mitochondrial dynamic impairment. These cardiometabolic modifications were accompanied by reduced c-kit+ and Sca-1+ cardiac progenitor cell (CPC) populations and impaired CPC differentiation into new cardiomyocytes. CONCLUSIONS In conclusion, Obese mother-offspring developed cardiometabolic changes in adulthood that negatively impacted the CPC niches and, consequently, the formation of new cardiomyocytes. This process seems to be an essential mechanism involved in the pathophysiology of the disease, impairing cardiac homeostasis.
Collapse
Affiliation(s)
- Daniela C Andrade
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil.
| | - Bruna Gaetani
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Sara Moura
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Simone N de Carvalho
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Alessandra A Thole
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Erika Cortez
- Stem Cell Research Laboratory, Institute of Biology, State University of Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Ghosh S, Ganguly A, Habib M, Shin BC, Thamotharan S, Andersson S, Devaskar SU. Hepatic and Pancreatic Cellular Response to Early Life Nutritional Mismatch. Endocrinology 2025; 166:bqaf007. [PMID: 39823439 PMCID: PMC11815087 DOI: 10.1210/endocr/bqaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/22/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
To determine the basis for perinatal nutritional mismatch causing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus, we examined adult phenotype, hepatic transcriptome, and pancreatic β-islet function. In prenatal caloric-restricted rats with intrauterine growth restriction (IUGR) and postnatal exposure to high fat with fructose (HFhf) or high carbohydrate, we investigated male and female IUGR-HFhf and IUGR-high carbohydrate, vs HFhf and control offspring. Males more than females displayed adiposity, glucose intolerance, insulin resistance, hyperlipidemia, and hepatomegaly with hepatic steatosis. Male hepatic triglyceride synthesis, de novo lipogenesis genes increased, while female lipolysis, β-oxidation, fatty acid efflux, and FGF21 genes increased. IUGR-HFhf males demonstrated reduced β-islet insulin and humanin, and type 1 diabetes mellitus human amniotic fluid increased humanin. Humanin suppression disabled glucose stimulated insulin, ATP production, with apoptotic diminished β-islet viability. Humanin and FGF21 may reverse perinatal nutritional mismatched phenotype by restoring functional β islets and preventing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus.
Collapse
Affiliation(s)
- Shubhamoy Ghosh
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Amit Ganguly
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Manal Habib
- Division of Endocrinology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Bo-Chul Shin
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Shanthie Thamotharan
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Sture Andersson
- Department of Pediatrics, Helsinki University Central Hospital, 00290 Helsinki, Finland
| | - Sherin U Devaskar
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| |
Collapse
|
8
|
Muntean M, Mărginean C, Bernad ES, Bănescu C, Nyulas V, Muntean IE, Săsăran V. The Link Between Newborn SNP Polymorphism rs266729, Adiponectin, and Newborn Macrosomia in a Cohort of Pregnant Women with Gestational Diabetes Mellitus: A Case-Control Pilot Study. CHILDREN (BASEL, SWITZERLAND) 2025; 12:155. [PMID: 40003257 PMCID: PMC11854160 DOI: 10.3390/children12020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/18/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is linked to higher newborn weight and an increased risk of macrosomia. The newborn single-nucleotide polymorphism (SNP) of the ADIPOQ gene rs266729 is linked to a higher birth weight of the offspring of healthy pregnant women. OBJECTIVES This study aims to evaluate the relationship between newborn ADIPOQ rs266729 polymorphism, cord blood adiponectin, maternal glycemic and lipid metabolism, and maternal adiponectin levels at 24 to 28 weeks of gestation (WG) and at birth and its impact on newborn weight in a cohort of GDM mothers. MATERIALS AND METHODS This study involved 71 women diagnosed with GDM and 142 control pregnant women. The ADIPOQ (rs266729) gene polymorphisms were genotyped using TaqMan real-time PCR analysis. Maternal and cord blood adiponectin levels were measured using human total adiponectin ELISA kits. We performed a Pearson correlation analysis to identify significant correlations between maternal metabolic parameters and adiponectin levels at 24-28 WG and birth and the weight of newborns. A logistic regression analysis was also conducted to identify potential macrosomia predictors. RESULTS We found no significant differences in the distribution of the allele (C, G) (p = 0.82) and genotype (CC, CG, GG) (p = 0.46) of APIPOQ rs266729 among normoponderal and macrosomic newborns from the GDM mothers group. Maternal fasting glucose at 24-28 WG was higher in the GDM mothers who gave birth to macrosomic newborns (106 ± 17 vs. 93 ± 10 mg/dL, p < 0.0001). Adiponectin levels in the cord blood of newborns from mothers with GDM were lower than those in newborns from control mothers (p < 0.0001). In correlation analysis, we identified a weak positive correlation between the newborn weight of GDM mothers and cord blood adiponectin (r = 0.262), maternal fasting glucose level at 24-28 WG (r = 0.288), and maternal adiponectin level at birth (0.334). Multivariate logistic regression, after adjusting for confounders, revealed that maternal fasting glucose levels at 24-28 WG had an OR of 11.59, and cord blood adiponectin levels had an OR of 30.31 for macrosomia. CONCLUSIONS The preliminary findings of our pilot study suggest that in the gestational diabetes mellitus group, the ADIPOQ rs266729 polymorphism in newborns is not associated with a higher birth weight, maternal fasting glucose levels between 24 and 28 WG were a predictor for macrosomia, and cord blood adiponectin levels were lower than those from control mothers. Further large-scale studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Mihai Muntean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Targu Mures, Romania; (M.M.); (V.S.)
| | - Claudiu Mărginean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Targu Mures, Romania; (M.M.); (V.S.)
| | - Elena Silvia Bernad
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
- Center for Laparoscopy, Laparoscopic Surgery and In Vitro Fertilization, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Claudia Bănescu
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Targu Mures, Romania;
| | - Victoria Nyulas
- Department of Informatics and Medical Biostatistics, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Targu Mures, Romania;
| | | | - Vladut Săsăran
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Targu Mures, Romania; (M.M.); (V.S.)
| |
Collapse
|
9
|
Aleksic V, Gazibara T, Jeremic B, Kulic L, Dotlic J, Stevanovic J, Milovanovic V, Milic M. Associations of night eating with body mass index and waist circumference among health sciences students living in a post-conflict region. Nutr Health 2025:2601060241309991. [PMID: 39828953 DOI: 10.1177/02601060241309991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND Stressors associated with the ongoing ethnic tensions may be the drivers of more frequent night eating, and subsequently, a higher body mass index. AIM To examine the association of night eating with body mass index (BMI) and waist circumference among health sciences students in Northern Kosovo. METHODS A cross-sectional study was carried out at the University of Prishtina in Kosovska Mitrovica from November 2018 to March 2019. Participants completed a socio-demographic questionnaire, the Night Eating Questionnaire (NEQ), the Beck's Anxiety Inventory (BAI) and the Perceived Stress Scale (PSS). Body weight, height, and waist circumference were measured by the research team. RESULTS The study sample consisted of 534 students. The night eating score was 14.4 ± 4.5 out of 52 (prevalence of night eating syndrome was 3.0%). Overall, the average BMI was 23.7 ± 4.5 kg/m2 (27.2% of students were overweight and 7.5% were obese). Moderate level of stress as measured by the PSS was observed 61.8% and high level of stress was recorded among 5.8%. Also, 9.2% students reported moderate BAI score and 6.0% students reported severe BAI score. After adjustment for socio-demographic characteristics, health behaviors, sleep, stress and anxiety levels, no association of night eating with BMI and waist circumference was observed. CONCLUSION These findings showed that there is an absence of the association between night eating, BMI and waist circumference among students who live in a post-conflict setting. There may be other factors that mediate the relationship between food intake at night and body mass index.
Collapse
Affiliation(s)
- Vojkan Aleksic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Secondary School for Nursing, Cuprija, Serbia
| | - Tatjana Gazibara
- Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Ljiljana Kulic
- Department of Occupational Medicine, Faculty of Medicine, University of Pristina temporarily seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Jelena Dotlic
- Clinic for Obstetrics and Gynecology, Clinical Center of Serbia, Belgrade, Serbia
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jasmina Stevanovic
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Vesna Milovanovic
- Department of Internal Medicine, Clinical Center of Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Marija Milic
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
- Department of Prevention, Institute of Public Health of Serbia "Dr Milan Jovanovic Batut", Belgrade, Serbia
| |
Collapse
|
10
|
Gyllenhammer LE, Boyle KE. New Frontiers: Umbilical Cord Mesenchymal Stem Cells Uncover Developmental Roots and Biological Underpinnings of Obesity Susceptibility. Curr Obes Rep 2025; 14:10. [PMID: 39814984 PMCID: PMC11735562 DOI: 10.1007/s13679-024-00599-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE OF REVIEW To review evidence supporting human umbilical cord mesenchymal stem cells (UC-MSC) as an innovative model system advancing obesity precision medicine. RECENT FINDINGS Obesity prevalence is increasing rapidly and exposures during fetal development can impact individual susceptibility to obesity. UC-MSCs exhibit heterogeneous phenotypes associated with maternal exposures and predictive of child cardiometabolic outcomes. This recent evidence supports UC-MSCs as a precision model serving three purposes: (1) as a mechanistic tool to interrogate biological underpinnings of obesity in human studies, (2) as a sensitive index of early life causes and determinants of obesity, and (3) as a marker and transducer of susceptibility, highlighting populations most at risk for future obesity. Data from UC-MSCs emphasize nutrient sensing and lipid partitioning as phenotypes most relevant to neonatal and early childhood adiposity and implicate a role for these cell-autonomous features of mesodermal tissues in the biological underpinnings of obesity.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, USA.
| | - Kristen E Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA.
| |
Collapse
|
11
|
Rakhshandehroo M, Harvey L, de Bruin A, Timmer E, Lohr J, Tims S, Schipper L. Maternal exposure to purified versus grain-based diet during early lactation in mice affects offspring growth and reduces responsivity to Western-style diet challenge in adulthood. J Dev Orig Health Dis 2025; 16:e3. [PMID: 39780545 DOI: 10.1017/s2040174424000436] [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: 01/11/2025]
Abstract
The nutritional environment during fetal and early postnatal life has a long-term impact on growth, development, and metabolic health of the offspring, a process termed "nutritional programming." Rodent models studying programming effects of nutritional interventions use either purified or grain-based rodent diets as background diets. However, the impact of these diets on phenotypic outcomes in these models has not been comprehensively investigated. We used a previously validated (C57BL/6J) mouse model to investigate the effects of infant milk formula (IMF) interventions on nutritional programming. Specifically, we investigated the effects of maternal diet type (i.e., grain-based vs purified) during early lactation and prior to the intervention on offspring growth, metabolic phenotype, and gut microbiota profile. Maternal exposure to purified diet led to an increased post-weaning growth velocity in the offspring and reduced adult diet-induced obesity. Further, maternal exposure to purified diet reduced the offspring gut microbiota diversity and modified its composition post-weaning. These data not only reinforce the notion that maternal nutrition significantly influences the programming of offspring vulnerability to an obesogenic diet in adulthood but emphasizes the importance of careful selection of standard background diet type when designing any preclinical study with (early life) nutritional interventions.
Collapse
Affiliation(s)
| | - L Harvey
- Danone Research & Innovation Center, Utrecht, The Netherlands
| | - A de Bruin
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - E Timmer
- Danone Research & Innovation Center, Utrecht, The Netherlands
| | - J Lohr
- Danone Research & Innovation Center, Utrecht, The Netherlands
| | - S Tims
- Danone Research & Innovation Center, Utrecht, The Netherlands
| | - L Schipper
- Danone Research & Innovation Center, Utrecht, The Netherlands
| |
Collapse
|
12
|
Ren J, Zhou L, Li S, Zhang Q, Xiao X. The roles of the gut microbiota, metabolites, and epigenetics in the effects of maternal exercise on offspring metabolism. Am J Physiol Endocrinol Metab 2024; 327:E760-E772. [PMID: 39535269 DOI: 10.1152/ajpendo.00200.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Metabolic diseases, including obesity, dyslipidemia, and type 2 diabetes, have become severe challenges worldwide. The Developmental Origins of Health and Disease (DOHaD) hypothesis suggests that an adverse intrauterine environment can increase the risk of metabolic disorders in offspring. Studies have demonstrated that maternal exercise is an effective intervention for improving the offspring metabolic health. However, the pathways through which exercise works are unclear. It has been reported that the gut microbiota mediates the effect of maternal exercise on offspring metabolism, and epigenetic modifications have also been proposed to be important molecular mechanisms. Microbial metabolites can influence epigenetics by providing substrates for DNA or histone modifications, binding to G-protein-coupled receptors to affect downstream pathways, or regulating the activity of epigenetic modifying enzymes. This review aims to summarize the intergenerational effect of maternal exercise and proposes that gut microbiota-metabolites-epigenetic regulation is an important mechanism by which maternal exercise improves offspring metabolism, which may yield novel targets for the early prevention and intervention of metabolic diseases.
Collapse
Affiliation(s)
- Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Łuszczki E, Wyszyńska J, Dymek A, Drożdż D, González-Ramos L, Hartgring I, García-Carbonell N, Mazur A, Erdine S, Parnarauskienė J, Alvarez-Pitti J. The Effect of Maternal Diet and Lifestyle on the Risk of Childhood Obesity. Metabolites 2024; 14:655. [PMID: 39728436 DOI: 10.3390/metabo14120655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Childhood obesity is a global health problem that affects at least 41 million children under the age of five. Increased BMI in children is associated with serious long-term health consequences, such as type 2 diabetes, cardiovascular disease, and psychological problems, including depression and low self-esteem. Although the etiology of obesity is complex, research suggests that the diet and lifestyle of pregnant women play a key role in shaping metabolic and epigenetic changes that can increase the risk of obesity in their children. Excessive gestational weight gain, unhealthy dietary patterns (including the Western diet), and pregnancy complications (such as gestational diabetes) are some of the modifiable factors that contribute to childhood obesity. The purpose of this narrative review is to summarize the most important and recent information on the impact of the diet and lifestyle of pregnant women on the risk of childhood obesity. Methods: This article is a narrative review that aims to summarize the available literature on the impact of pregnant women's diet and lifestyle on the risk of obesity in their offspring, with a focus on metabolic and epigenetic mechanisms. Results/Conclusions: Current evidence suggests that a pregnant woman's lifestyle and diet can significantly contribute to lowering the risk of obesity in their offspring. However, further high-quality research is needed to understand better the metabolic and epigenetic relationships concerning maternal factors that predispose offspring to obesity.
Collapse
Affiliation(s)
- Edyta Łuszczki
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Justyna Wyszyńska
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Dymek
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Pediatric Institute, Jagiellonian University Medical College, 31-007 Krakow, Poland
| | - Laura González-Ramos
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Isa Hartgring
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Nuria García-Carbonell
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
| | - Artur Mazur
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Serap Erdine
- Cerrahpasa Faculty of Medicine, Department of Cardiology, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
| | - Justė Parnarauskienė
- Pediatric Department, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Julio Alvarez-Pitti
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Quarta A, Quarta MT, Mastromauro C, Chiarelli F, Giannini C. Influence of Nutrition on Growth and Development of Metabolic Syndrome in Children. Nutrients 2024; 16:3801. [PMID: 39599588 PMCID: PMC11597107 DOI: 10.3390/nu16223801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Obesity is currently an increasing public health burden due to its related metabolic and cardiovascular complications. In Western countries, a significant number of people are overweight or obese, and this trend is, unfortunately, becoming increasingly common even among the pediatric population. In this narrative review, we analyzed the role of nutrition during growth and its impact on the risk of developing metabolic syndrome and cardiovascular complications later in life. An impactful role in determining the phenotypic characteristics of the offspring is the parental diet carried out before conception. During intrauterine growth, the main risk factors are represented by an unbalanced maternal diet, excessive gestational weight gain, and impaired glycemic status. Breastfeeding, on the other hand, has many beneficial effects, but at the same time the quality of breast milk may be modified if maternal overweight or obesity subsists. Complementary feeding is likewise pivotal because an early introduction before 4 months of age and a high protein intake contribute to weight gain later. Knowledge of these mechanisms may allow early modification of risk factors by implementing targeted preventive strategies.
Collapse
Affiliation(s)
| | | | | | | | - Cosimo Giannini
- Department of Pediatrics, University of Chieti—Pescara, G. D’Annunzio, 66100 Chieti, Italy; (A.Q.); (M.T.Q.); (C.M.); (F.C.)
| |
Collapse
|
15
|
Waldrop SW, Perng W, Konigsberg IR, Borengasser SJ. The potential utility of cord blood DNA methylation in pediatric clinical practice. Epigenomics 2024; 16:1365-1372. [PMID: 39530586 PMCID: PMC11622741 DOI: 10.1080/17501911.2024.2408217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
Our understanding of the origins of noncommunicable diseases has evolved over the years with greater consideration given to the lasting influence exposures and experiences during the preconceptional and prenatal periods can have. Research highlights the associations of parental exposures (e.g., diet, obesity, gestational diabetes, lipid profile, toxic exposures and microbiome) with the infant/fetal methylome and suggest associations with infant, child and/or adolescent chronic health outcomes. Thus, epigenetics and specifically cord blood DNA methylation may have utility as biomarkers for disease risk identification and stratification in pediatrics. However, for cord blood DNA methylation analyses to be leveraged as biomarkers of disease risk in pediatric clinical practice, the results must be replicable, validated and clinically meaningful. Challenges and opportunities to this prospect are herein discussed.
Collapse
Affiliation(s)
- Stephanie W Waldrop
- Section on Nutrition, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO80045, USA
- Division of Clinical Sciences, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA70808, USA
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity & Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO80045, USA
| | - Iain R Konigsberg
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO80045, USA
| | - Sarah J Borengasser
- Department of Pediatrics, TSET Health Promotion Research Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104, USA
| |
Collapse
|
16
|
Gems D, Virk RS, de Magalhães JP. Epigenetic clocks and programmatic aging. Ageing Res Rev 2024; 101:102546. [PMID: 39414120 DOI: 10.1016/j.arr.2024.102546] [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: 07/24/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
The last decade has seen remarkable progress in the characterization of methylation clocks that can serve as indicators of biological age in humans and many other mammalian species. While the biological processes of aging that underlie these clocks have remained unclear, several clues have pointed to a link to developmental mechanisms. These include the presence in the vicinity of clock CpG sites of genes that specify development, including those of the Hox (homeobox) and polycomb classes. Here we discuss how recent advances in programmatic theories of aging provide a framework within which methylation clocks can be understood as part of a developmental process of aging. This includes how such clocks evolve, how developmental mechanisms cause aging, and how they give rise to late-life disease. The combination of ideas from evolutionary biology, biogerontology and developmental biology open a path to a new discipline, that of developmental gerontology (devo-gero). Drawing on the properties of methylation clocks, we offer several new hypotheses that exemplify devo-gero thinking. We suggest that polycomb controls a trade-off between earlier developmental fidelity and later developmental plasticity. We also propose the existence of an evolutionarily-conserved developmental sequence spanning ontogenesis, adult development and aging, that both constrains and determines the evolution of aging.
Collapse
Affiliation(s)
- David Gems
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom.
| | - Roop Singh Virk
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - João Pedro de Magalhães
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, B15 2WB, United Kingdom
| |
Collapse
|
17
|
Li Y, Yu X, Shi J, Zhao J, Li L. The role of aryl hydrocarbon receptors in nutrient metabolism and immune regulation at the maternal-fetal interface. Placenta 2024; 154:9-17. [PMID: 38830294 DOI: 10.1016/j.placenta.2024.05.134] [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: 10/18/2023] [Revised: 04/29/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
The maternal-fetal interface is composed of the placenta, which is affiliated with the fetus, and the maternal decidua. During pregnancy, the placenta is mainly responsible for nutrient transport and immune tolerance maintenance, which plays a key role in fetal growth and development and pregnancy maintenance. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that exists in various cell types at the maternal-fetal interface and is involved in multiple cellular processes. Recent studies have highlighted the role of AhR in regulating various physiological processes, including glucose and lipid metabolism, as well as tryptophan metabolism and immune responses, within non-pregnant tissues. This review shifts focus towards understanding how AhR modulation impacts metabolism and immune regulation at the maternal-fetal interface. This may implicate the development of pregnancy-related complications and the potential target of the AhR pathway for therapeutic strategies against poor pregnancy outcomes.
Collapse
Affiliation(s)
- Yuchen Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, Shandong, 250021, China
| | - Xiaojun Yu
- School of Public Health Kunming Medical University, Kunming, 650500, China
| | - Jing Shi
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
| | - Jie Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100083, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100083, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100083, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100083, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
18
|
He H, Chen J, Hua Y, Xie Z, Tu M, Liu L, Wang H, Yang X, Chen L. α7-nAChR/P300/NLRP3-regulated pyroptosis mediated poor articular cartilage quality induced by prenatal nicotine exposure in female offspring rats. Chem Biol Interact 2024; 400:111183. [PMID: 39098741 DOI: 10.1016/j.cbi.2024.111183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Nicotine is developmentally toxic. Prenatal nicotine exposure (PNE) affects the development of multiple fetal organs and causes susceptibility to a variety of diseases in offspring. In this study, we aimed to investigate the effect of PNE on cartilage development and osteoarthritis susceptibility in female offspring rats. Wistar rats were orally gavaged with nicotine on days 9-20 of pregnancy. The articular cartilage was obtained at gestational day (GD) 20 and postnatal week (PW) 24, respectively. Further, the effect of nicotine on chondrogenic differentiation was explored by the chondrogenic differentiation model in human Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs). The PNE group showed significantly shallower Safranin O staining and lower Collagen 2a1 content of articular cartilage in female offspring rats. Further, we found that PNE activated pyroptosis in the articular cartilage at GD20 and PW24. In vitro experiments revealed that nicotine inhibited chondrogenic differentiation and activated pyroptosis. After interfering with nod-like receptors3 (NLRP3) expression by SiRNA, it was found that pyroptosis mediated the chondrogenic differentiation inhibition of WJ-MSCs induced by nicotine. In addition, we found that α7-nAChR antagonist α-BTX reversed nicotine-induced NLRP3 and P300 high expression. And, P300 SiRNA reversed the increase of NLRP3 mRNA expression and histone acetylation level in its promoter region induced by nicotine. In conclusion, PNE caused chondrodysplasia and poor articular cartilage quality in female offspring rats. PNE increased the histone acetylation level of NLRP3 promoter region by α7-nAChR/P300, which resulting in the high expression of NLRP3. Further, NLRP3 mediated the inhibition of chondrogenic differentiation by activating pyroptosis.
Collapse
Affiliation(s)
- Hangyuan He
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yi Hua
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhe Xie
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Tu
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Liang Liu
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Xu Yang
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liaobin Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
19
|
Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, Simcox J, Susztak K, Drucker DJ. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell 2024; 187:3789-3820. [PMID: 39059357 PMCID: PMC11299851 DOI: 10.1016/j.cell.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Diabetes, a complex multisystem metabolic disorder characterized by hyperglycemia, leads to complications that reduce quality of life and increase mortality. Diabetes pathophysiology includes dysfunction of beta cells, adipose tissue, skeletal muscle, and liver. Type 1 diabetes (T1D) results from immune-mediated beta cell destruction. The more prevalent type 2 diabetes (T2D) is a heterogeneous disorder characterized by varying degrees of beta cell dysfunction in concert with insulin resistance. The strong association between obesity and T2D involves pathways regulated by the central nervous system governing food intake and energy expenditure, integrating inputs from peripheral organs and the environment. The risk of developing diabetes or its complications represents interactions between genetic susceptibility and environmental factors, including the availability of nutritious food and other social determinants of health. This perspective reviews recent advances in understanding the pathophysiology and treatment of diabetes and its complications, which could alter the course of this prevalent disorder.
Collapse
Affiliation(s)
- E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Department of Genetics, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, and Imperial College NHS Trust, London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Ding L, Weger BD, Liu J, Zhou L, Lim Y, Wang D, Xie Z, Liu J, Ren J, Zheng J, Zhang Q, Yu M, Weger M, Morrison M, Xiao X, Gachon F. Maternal high fat diet induces circadian clock-independent endocrine alterations impacting the metabolism of the offspring. iScience 2024; 27:110343. [PMID: 39045103 PMCID: PMC11263959 DOI: 10.1016/j.isci.2024.110343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Maternal obesity has long-term effects on offspring metabolic health. Among the potential mechanisms, prior research has indicated potential disruptions in circadian rhythms and gut microbiota in the offspring. To challenge this hypothesis, we implemented a maternal high fat diet regimen before and during pregnancy, followed by a standard diet after birth. Our findings confirm that maternal obesity impacts offspring birth weight and glucose and lipid metabolisms. However, we found minimal impact on circadian rhythms and microbiota that are predominantly driven by the feeding/fasting cycle. Notably, maternal obesity altered rhythmic liver gene expression, affecting mitochondrial function and inflammatory response without disrupting the hepatic circadian clock. These changes could be explained by a masculinization of liver gene expression similar to the changes observed in polycystic ovarian syndrome. Intriguingly, such alterations seem to provide the first-generation offspring with a degree of protection against obesity when exposed to a high fat diet.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Benjamin D. Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100022, China
| | - Yenkai Lim
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Dongmei Wang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ziyan Xie
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Miao Yu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Meltem Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Mark Morrison
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Australian Infectious Diseases Research Centre, St. Lucia, QLD 4072, Australia
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Frédéric Gachon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
21
|
Sayeed UB, Akhtar E, Roy AK, Akter S, von Ehrenstein OS, Raqib R, Wagatsuma Y. Fetal femur length and risk of diabetes in adolescence: a prospective cohort study. Trop Med Health 2024; 52:44. [PMID: 38951934 PMCID: PMC11218328 DOI: 10.1186/s41182-024-00611-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Diabetes is more apparent in adulthood but may be dormant in childhood and originates during early fetal development. In fetal biometry, femur length (FL) is crucial for assessing fetal growth and development. This study aimed to assess potential associations between fetal femur growth and prediabetic biomarkers in Bangladeshi children. METHODS A cohort study embedded in a population-based maternal food and micronutrient supplementation (MINIMat) trial was conducted in Matlab, Bangladesh. The children in the cohort were followed up until 15 years of age. In the original trial, pregnancy was confirmed by ultrasound before 13 gestational weeks (GWs). Afterward, ultrasound assessments were performed at 14, 19, and 30 GWs. FL was measured from one end to the other, capturing a complete femoral image. The FL was standardized by GW, and a z-score was calculated. FBG and HbA1c levels were determined in plasma and whole blood, and the triglyceride-glucose index, a biomarker of insulin resistance, was calculated as Ln [fasting triglycerides (mg/dl) × fasting glucose (mg/dl)/2]. Multivariable linear regression analysis using a generalized linear model was performed to estimate the effects of FL at 14, 19 and 30 GWs on prediabetic biomarkers at 9 and 15 years of age. Maternal micronutrient and food supplementation group, parity, child sex, and BMI at 9 years or 15 years were included as covariates. RESULTS A total of 1.2% (6/515) of the participants had impaired fasting glucose during preadolescence, which increased to 3.5% (15/433) during adolescence. At 9 years, 6.3% (32/508) of the participants had elevated HbA1c%, which increased to 28% (120/431) at 15 years. Additionally, the TyG index increased from 9.5% (49/515) (during preadolescence) to 13% (56/433) (during adolescence). A one standard deviation decrease in FL at 14 and 19 GWs was associated with increased FBG (β = - 0.44 [- 0.88, - 0.004], P = 0.048; β = - 0.59 [- 1.12, - 0.05], P = 0.031) and HbA1c (β = - 0.01; [- 0.03, -0.005], P = 0.007; β = - 0.01 [- 0.03, - 0.003], P = 0.018) levels at 15 years. FL was not associated with diabetic biomarkers at 9 years. CONCLUSION Mid-trimester impaired femur growth may be associated with elevated prediabetic biomarkers in Bangladeshi adolescents.
Collapse
Affiliation(s)
- Urme Binte Sayeed
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Evana Akhtar
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Anjan Kumar Roy
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Sharmin Akter
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Ondine S von Ehrenstein
- Departments of Community Health Sciences and Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Rubhana Raqib
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Yukiko Wagatsuma
- Faculty of Medicine, Department of Clinical Trials and Clinical Epidemiology, University of Tsukuba, Tsukuba, Ibaraki, 805-3575, Japan
| |
Collapse
|
22
|
Shi H, Yang D, Ma L, Cheng Y, Liu Y, Ma J, Tong H, Shi C. Early life malnutrition and risk of T2DM adulthood: evidence from the lower socioeconomic status of northwest Chinese population. Front Nutr 2024; 11:1379725. [PMID: 38993241 PMCID: PMC11236714 DOI: 10.3389/fnut.2024.1379725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
Objective This study aimed to explore whether famine exposure during early life are associated with a high risk of Type 2 Diabetes Mellitus (T2DM) in adulthood and the role of socioeconomic status (SES) on this effect. Materials and methods We conducted a secondary data analysis based on data from a cross-sectional survey, collected 3,355 participants born between January 1, 1941 and December 31, 1966. Participants were categorized into four groups based on their date of birth, unexposed (individuals born in 1963-1966), infant exposed (individuals born in 1959-1962), childhood exposed (individuals born in 1949-1958), and adolescent exposed (born in 1941-1948). The association of famine exposure with T2DM risk in adults and conducted separately in plain area and mountain area was assessed using logistics regression model. Result 22.35% of participants were diagnosed with T2DM, of which 43.47% were from the childhood famine-exposed group, representing the highest proportion among all subgroups (p < 0.001). Participants exposed to famine during childhood and adolescence from the lower SES mountain areas showed a significantly higher prevalence of T2DM in adulthood than those from the plain areas (p < 0.001). The adolescence stage exposed famine will increase the risk of T2DM in the mountain area (OR 2.46, 95% CI 1.61, 3.77). Conclusion No strong evidence demonstrates that exposure to famine during the early life stage increases the risk of developing T2DM in adulthood. However, populations with lower SES are likely to be exposed to more risk factors for T2DM.
Collapse
Affiliation(s)
- Hongjuan Shi
- School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Danyu Yang
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Institute, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Ling Ma
- School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yin Cheng
- School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yining Liu
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Institute, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jinyu Ma
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Institute, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Huitian Tong
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Institute, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Chao Shi
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Institute, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| |
Collapse
|
23
|
Heinecke F, Fornes D, Capobianco E, Flores Quiroga JP, Labiano M, Faletti AG, Jawerbaum A, White V. Intestinal alterations and mild glucose homeostasis impairments in the offspring born to overweight rats. Mol Cell Endocrinol 2024; 587:112201. [PMID: 38494045 DOI: 10.1016/j.mce.2024.112201] [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: 12/29/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
The gut plays a crucial role in metabolism by regulating the passage of nutrients, water and microbial-derived substances to the portal circulation. Additionally, it produces incretins, such as glucose-insulinotropic releasing peptide (GIP) and glucagon-like derived peptide 1 (GLP1, encoded by gcg gene) in response to nutrient uptake. We aimed to investigate whether offspring from overweight rats develop anomalies in the barrier function and incretin transcription. We observed pro-inflammatory related changes along with a reduction in Claudin-3 levels resulting in increased gut-permeability in fetuses and offspring from overweight rats. Importantly, we found decreased gip mRNA levels in both fetuses and offspring from overweight rats. Differently, gcg mRNA levels were upregulated in fetuses, downregulated in female offspring and unchanged in male offspring from overweight rats. When cultured with high glucose, intestinal explants showed an increase in gip and gcg mRNA levels in control offspring. In contrast, offspring from overweight rats did not exhibit any response in gip mRNA levels. Additionally, while females showed no response, male offspring from overweight rats did exhibit an upregulation in gcg mRNA levels. Furthermore, female and male offspring from overweight rats showed sex-dependent anomalies when orally challenged with a glucose overload, returning to baseline glucose levels after 120 min. These results open new research questions about the role of the adverse maternal metabolic condition in the programming of impairments in glucose homeostasis, enteroendocrine function and gut barrier function in the offspring from overweight mothers and highlight the importance of a perinatal maternal healthy metabolism.
Collapse
Affiliation(s)
- Florencia Heinecke
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Daiana Fornes
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Jeremias Pablo Flores Quiroga
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Marina Labiano
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia G Faletti
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Verónica White
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Sindhu P, Magotra A, Sindhu V, Chaudhary P. Unravelling the impact of epigenetic mechanisms on offspring growth, production, reproduction and disease susceptibility. ZYGOTE 2024; 32:190-206. [PMID: 39291610 DOI: 10.1017/s0967199424000224] [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: 09/19/2024]
Abstract
Epigenetic mechanisms, such as DNA methylation, histone modifications and non-coding RNA molecules, play a critical role in gene expression and regulation in livestock species, influencing development, reproduction and disease resistance. DNA methylation patterns silence gene expression by blocking transcription factor binding, while histone modifications alter chromatin structure and affect DNA accessibility. Livestock-specific histone modifications contribute to gene expression and genome stability. Non-coding RNAs, including miRNAs, piRNAs, siRNAs, snoRNAs, lncRNAs and circRNAs, regulate gene expression post-transcriptionally. Transgenerational epigenetic inheritance occurs in livestock, with environmental factors impacting epigenetic modifications and phenotypic traits across generations. Epigenetic regulation revealed significant effect on gene expression profiling that can be exploited for various targeted traits like muscle hypertrophy, puberty onset, growth, metabolism, disease resistance and milk production in livestock and poultry breeds. Epigenetic regulation of imprinted genes affects cattle growth and metabolism while epigenetic modifications play a role in disease resistance and mastitis in dairy cattle, as well as milk protein gene regulation during lactation. Nutri-epigenomics research also reveals the influence of maternal nutrition on offspring's epigenetic regulation of metabolic homeostasis in cattle, sheep, goat and poultry. Integrating cyto-genomics approaches enhances understanding of epigenetic mechanisms in livestock breeding, providing insights into chromosomal structure, rearrangements and their impact on gene regulation and phenotypic traits. This review presents potential research areas to enhance production potential and deepen our understanding of epigenetic changes in livestock, offering opportunities for genetic improvement, reproductive management, disease control and milk production in diverse livestock species.
Collapse
Affiliation(s)
- Pushpa Sindhu
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Ankit Magotra
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Vikas Sindhu
- Department of Animal Nutrition, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Pradeep Chaudhary
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
25
|
Dearden L, Furigo IC, Pantaleão LC, Wong LWP, Fernandez-Twinn DS, de Almeida-Faria J, Kentistou KA, Carreira MV, Bidault G, Vidal-Puig A, Ong KK, Perry JRB, Donato J, Ozanne SE. Maternal obesity increases hypothalamic miR-505-5p expression in mouse offspring leading to altered fatty acid sensing and increased intake of high-fat food. PLoS Biol 2024; 22:e3002641. [PMID: 38833481 PMCID: PMC11149872 DOI: 10.1371/journal.pbio.3002641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
In utero exposure to maternal obesity programs increased obesity risk. Animal models show that programmed offspring obesity is preceded by hyperphagia, but the mechanisms that mediate these changes are unknown. Using a mouse model of maternal obesity, we observed increased intake of a high-fat diet (HFD) in offspring of obese mothers that precedes the development of obesity. Through small RNA sequencing, we identified programmed overexpression of hypothalamic miR-505-5p that is established in the fetus, lasts to adulthood and is maintained in hypothalamic neural progenitor cells cultured in vitro. Metabolic hormones and long-chain fatty acids associated with obesity increase miR-505-5p expression in hypothalamic neurons in vitro. We demonstrate that targets of miR-505-5p are enriched in fatty acid metabolism pathways and overexpression of miR-505-5p decreased neuronal fatty acid metabolism in vitro. miR-505-5p targets are associated with increased BMI in human genetic studies. Intra-cerebroventricular injection of miR-505-5p in wild-type mice increased HFD intake, mimicking the phenotype observed in offspring exposed to maternal obesity. Conversely, maternal exercise intervention in an obese mouse pregnancy rescued the programmed increase of hypothalamic miR-505-5p in offspring of obese dams and reduced HFD intake to control offspring levels. This study identifies a novel mechanism by which maternal obesity programs obesity in offspring via increased intake of high-fat foods.
Collapse
Affiliation(s)
- Laura Dearden
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Isadora C. Furigo
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo, Brazil
- Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Lucas C. Pantaleão
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - L W. P. Wong
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Denise S. Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Juliana de Almeida-Faria
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
- University of Campinas, Faculty of Medical Sciences, Department of Pharmacology, Campinas, Brazil
| | | | - Maria V. Carreira
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Ken K. Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - John R. B. Perry
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo, Brazil
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| |
Collapse
|
26
|
Toledano JM, Puche-Juarez M, Moreno-Fernandez J, Gonzalez-Palacios P, Rivas A, Ochoa JJ, Diaz-Castro J. Implications of Prenatal Exposure to Endocrine-Disrupting Chemicals in Offspring Development: A Narrative Review. Nutrients 2024; 16:1556. [PMID: 38892490 PMCID: PMC11173790 DOI: 10.3390/nu16111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
During the last decades, endocrine-disrupting chemicals (EDCs) have attracted the attention of the scientific community, as a result of a deepened understanding of their effects on human health. These compounds, which can reach populations through the food chain and a number of daily life products, are known to modify the activity of the endocrine system. Regarding vulnerable groups like pregnant mothers, the potential damage they can cause increases their importance, since it is the health of two lives that is at risk. EDCs can affect the gestation process, altering fetal development, and eventually inducing the appearance of many disorders in their childhood and/or adulthood. Because of this, several of these substances have been studied to clarify the influence of their prenatal exposure on the cognitive and psychomotor development of the newborn, together with the appearance of non-communicable diseases and other disorders. The most novel research on the subject has been gathered in this narrative review, with the aim of clarifying the current knowledge on the subject. EDCs have shown, through different studies involving both animal and human investigation, a detrimental effect on the development of children exposed to the during pregnancy, sometimes with sex-specific outcomes. However, some other studies have failed to find these associations, which highlights the need for deeper and more rigorous research, that will provide an even more solid foundation for the establishment of policies against the extended use of these chemicals.
Collapse
Affiliation(s)
- Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.J.O.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Maria Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.J.O.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.J.O.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain;
| | - Patricia Gonzalez-Palacios
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Department of Nutrition and Food Science, University of Granada, 18071 Granada, Spain
| | - Ana Rivas
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain;
- Department of Nutrition and Food Science, University of Granada, 18071 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.J.O.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain;
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.J.O.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain;
| |
Collapse
|
27
|
Nicolaou M, Toumba M, Kythreotis A, Daher H, Skordis N. Obesogens in Adolescence: Challenging Aspects and Prevention Strategies. CHILDREN (BASEL, SWITZERLAND) 2024; 11:602. [PMID: 38790597 PMCID: PMC11120186 DOI: 10.3390/children11050602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Childhood obesity has become a global epidemic, with significant increases in prevalence over recent decades. While excessive calorie consumption and physical inactivity are known factors, emerging research highlights the role of endocrine-disrupting chemicals (EDCs), particularly obesogens, in obesity's pathogenesis. This review explores the historical context of the environmental obesogens hypothesis, their sources, mechanism of action, impact on prenatal and postnatal development, and epigenetics. Additionally, it discusses the long-term consequences of childhood obesity and proposes prevention strategies that will mitigate negative health effects. Obesogens were found to disrupt hormonal balance and metabolic processes through various mechanisms such as altering gene expression, hormonal interference, and inflammation. Especially significant was exposure during critical windows of development, which correlates with an increased risk of obesity in childhood or adolescence. Long-term effects of childhood obesity include chronic health conditions and psychosocial issues. A comprehensive approach is necessary to address childhood obesity encompassing genetic, environmental, and lifestyle factors. Prevention strategies should focus on reducing obesogen exposure, promoting healthy lifestyles, and implementing regulatory policies. Future research should investigate obesogens-diet interactions, microbiome impacts, and combined obesogens effects. Long-term human studies are also crucial for validating findings from animal models and allowing for informed decision-making to combat the obesity pandemic.
Collapse
Affiliation(s)
- Marina Nicolaou
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK; (M.N.); (A.K.)
| | - Meropi Toumba
- Paediatric Endocrinology Clinic, Department of Paediatrics, Aretaeio Hospital, 2024 Nicosia, Cyprus;
- School of Medicine, University of Nicosia, 2414 Nicosia, Cyprus;
| | - Alexandros Kythreotis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK; (M.N.); (A.K.)
| | - Habib Daher
- School of Medicine, University of Nicosia, 2414 Nicosia, Cyprus;
| | - Nicos Skordis
- School of Medicine, University of Nicosia, 2414 Nicosia, Cyprus;
- Division of Paediatric Endocrinology, Paedi Center for Specialized Paediatrics, 2025 Nicosia, Cyprus
| |
Collapse
|
28
|
Sheu WH. Birthweight and risk of diabetes in later life. J Diabetes Investig 2024; 15:557-558. [PMID: 38279766 PMCID: PMC11060154 DOI: 10.1111/jdi.14154] [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: 12/24/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/28/2024] Open
Abstract
Compared with normative birthweight persons, individuals with a lower birthweight had a higher chance of developing type 2 diabetes at a younger age, a lower body mass index at diagnosis, a lesser family history of diabetes and greater probability of comorbidities.
Collapse
Affiliation(s)
- Wayne Huey‐Herng Sheu
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanTaiwan
- Division of Endocrinology and Metabolism, Department of Internal MedicineTaichung Veterans General HospitalTaichungTaiwan
- Division of Endocrinology and Metabolism, Department of Internal MedicineTaipei Veterans General HospitalTaipeiTaiwan
| |
Collapse
|
29
|
Saavedra LPJ, Piovan S, Moreira VM, Gonçalves GD, Ferreira ARO, Ribeiro MVG, Peres MNC, Almeida DL, Raposo SR, da Silva MC, Barbosa LF, de Freitas Mathias PC. Epigenetic programming for obesity and noncommunicable disease: From womb to tomb. Rev Endocr Metab Disord 2024; 25:309-324. [PMID: 38040983 DOI: 10.1007/s11154-023-09854-w] [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] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Several epidemiological, clinical and experimental studies in recent decades have shown the relationship between exposure to stressors during development and health outcomes later in life. The characterization of these susceptible phases, such as preconception, gestation, lactation and adolescence, and the understanding of factors that influence the risk of an adult individual for developing obesity, metabolic and cardiovascular diseases, is the focus of the DOHaD (Developmental Origins of Health and Disease) research line. In this sense, advancements in molecular biology techniques have contributed significantly to the understanding of the mechanisms underlying the observed phenotypes, their morphological and physiological alterations, having as a main driving factor the epigenetic modifications and their consequent modulation of gene expression. The present narrative review aimed to characterize the different susceptible phases of development and associated epigenetic modifications, and their implication in the development of non-communicable diseases. Additionally, we provide useful insights into interventions during development to counteract or prevent long-term programming for disease susceptibility.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maria Natália Chimirri Peres
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Scarlett Rodrigues Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Mariane Carneiro da Silva
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Letícia Ferreira Barbosa
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil.
| |
Collapse
|
30
|
Martins MG, Woodside B, Kiss ACI. Effects of maternal mild hyperglycemia associated with snack intake on offspring metabolism and behavior across the lifespan. Physiol Behav 2024; 276:114483. [PMID: 38331375 DOI: 10.1016/j.physbeh.2024.114483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
The increasing prevalence of diabetes is of particular concern in women of childbearing age because of the short and long-term consequences of maternal diabetes for the health of the offspring, such as a greater risk of developing metabolic impairments and cognitive deficits. In addition, maternal diet during pregnancy and lactation might contribute to preventing or ameliorating adverse offspring outcomes. Recently, we described that access to snacks exacerbates glucose intolerance in mildly hyperglycemic pregnant dams. Therefore, we hypothesized that these offspring would show greater impairment in metabolic and behavioral outcomes across the lifespan. Neonatal STZ treatment was employed to induce maternal mild hyperglycemia in females. After mating, normo- and hyperglycemic dams were given access either to standard chow or standard show plus snacks. Male and female offspring were evaluated on postnatal days (PND) 30, 90, and 360. Offspring behavior was assessed in the marble burying task, the open-field test, the elevated-plus maze, and sucrose preference. Glucose tolerance and morphometric analyses were also carried out. Maternal hyperglycemia increased body weight and fat deposition only on PND 30, while retroperitoneal fat deposition was reduced in the offspring of snack-fed dams. However, maternal snack intake reduced offspring body weight and length on PND 90. Fasting glucose was increased in females born to hyperglycemic, snack-fed dams on PND 90. Glucose clearance was altered by both maternal conditions in male offspring on PND 30, however, this sex difference was reversed on PND 90, with maternal hyperglycemia impairing glucose clearance only in females. In addition, maternal hyperglycemia reduced anxiety-like behavior in female offspring on PND 30, especially in the offspring of snack-fed dams, while maternal snack intake reduced sucrose preference in both males and females in adulthood. These results suggest that the effects of maternal hyperglycemia during pregnancy and lactation on offspring outcomes were not exacerbated by snack intake. Although additive effects of the two maternal conditions were hypothesized, the absence of such effects could be related to the mild maternal hyperglycemia induced by STZ treatment even when combined with snack intake. While maternal hyperglycemia alone impaired some offspring outcomes, its association with snack intake did not aggravate those impairments but rather resulted in outcomes more similar to those of offspring born to normoglycemic dams. Finally, females were found to be more susceptible to both the effects of maternal hyperglycemia and snack intake on metabolism and behavior.
Collapse
Affiliation(s)
- Marina Galleazzo Martins
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), Rua do Matão, trav. 14, 321, Cidade Universitária, São Paulo, 05508-090, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Department of Structural and Functional Biology, Rua Prof. Dr. Antonio Celso Wagner Zanin, s/n, Botucatu, São Paulo, 18618-689, Brazil.
| | - Barbara Woodside
- Center for Studies in Behavioral Neurobiology, Psychology Department, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Ana Carolina Inhasz Kiss
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), Rua do Matão, trav. 14, 321, Cidade Universitária, São Paulo, 05508-090, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Department of Structural and Functional Biology, Rua Prof. Dr. Antonio Celso Wagner Zanin, s/n, Botucatu, São Paulo, 18618-689, Brazil
| |
Collapse
|
31
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [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: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
32
|
Burton JJN, Alonso LC. Overnutrition in the early postnatal period influences lifetime metabolic risk: Evidence for impact on pancreatic β-cell mass and function. J Diabetes Investig 2024; 15:263-274. [PMID: 38193815 PMCID: PMC10906026 DOI: 10.1111/jdi.14136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Overconsumption of energy-rich foods that disrupt caloric balance is a fundamental cause of overweight, obesity and diabetes. Dysglycemia and the resulting cardiovascular disease cause substantial morbidity and mortality worldwide, as well as high societal cost. The prevalence of obesity in childhood and adolescence is increasing, leading to younger diabetes diagnosis, and higher severity of microvascular and macrovascular complications. An important goal is to identify early life conditions that increase future metabolic risk, toward the goal of preventing diabetes and cardiovascular disease. An ample body of evidence implicates prenatal and postnatal childhood growth trajectories in the programming of adult metabolic disease. Human epidemiological data show that accelerated childhood growth increases risk of type 2 diabetes in adulthood. Type 2 diabetes results from the combination of insulin resistance and pancreatic β-cell failure, but specific mechanisms by which accelerated postnatal growth impact one or both of these processes remain uncertain. This review explores the metabolic impact of overnutrition during postnatal life in humans and in rodent models, with specific attention to the connection between accelerated childhood growth and future adiposity, insulin resistance, β-cell mass and β-cell dysfunction. With improved knowledge in this area, we might one day be able to modulate nutrition and growth in the critical postnatal window to maximize lifelong metabolic health.
Collapse
Affiliation(s)
- Joshua JN Burton
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health, Weill Cornell MedicineNew York CityNew YorkUSA
| | - Laura C Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health, Weill Cornell MedicineNew York CityNew YorkUSA
| |
Collapse
|
33
|
Wang X, He C, Wu N, Tian Y, An S, Chen W, Liu X, Zhang H, Xiong S, Liu Y, Li Q, Zhou Y, Shen X. Establishment and validation of a prediction model for gestational diabetes. Diabetes Obes Metab 2024; 26:663-672. [PMID: 38073424 DOI: 10.1111/dom.15356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024]
Abstract
AIM To develop a visual prediction model for gestational diabetes (GD) in pregnant women and to establish an effective and practical tool for clinical application. METHODS To establish a prediction model, the modelling set included 1756 women enrolled in the Zunyi birth cohort, the internal validation set included 1234 enrolled women, and pregnant women in the Wuhan cohort were included in the external validation set. We established a demographic-lifestyle factor model (DLFM) and a demographic-lifestyle-environmental pollution factor model (DLEFM) based on whether the women were exposed to environmental pollutants. The least absolute shrinkage and selection lasso-logistic regression analyses were used to identify the independent predictors of GD and construct a nomogram for predicting its occurrence. RESULTS The DLEFM regression analysis showed that a family history of diabetes (odd ratio [OR] 2.28; 95% confidence interval [CI] 1.05-4.71), a history of GD in pregnant women (OR 4.22; 95% CI 1.89-9.41), being overweight or obese before pregnancy (OR 1.71; 95% CI 1.27-2.29), a history of hypertension (OR 2.61; 95% CI 1.41-4.72), sedentary time (h/day) (OR 1.16; 95% CI 1.08-1.24), monobenzyl phthalate (OR 1.95; 95% CI 1.45-2.67) and Q4 mono-ethyl phthalate concentration (OR 1.85; 95% CI 1.26-2.73) were independent predictors. The area under the receiver operating curves for the internal validation of the DLEFM and the DLFM constructed using these seven factors was 0.827 and 0.783, respectively. The calibration curve of the DLEFM was close to the diagonal line. The DLEFM was thus the more optimal model, and the one which we chose. CONCLUSIONS A nomogram based on preconception factors was constructed to predict the occurrence of GD in the second and third trimesters. It provided an effective tool for the early prediction and timely management of GD.
Collapse
Affiliation(s)
- Xia Wang
- School of Public Health, Zunyi Medical University, Zunyi, China
- Department of Non-Communicable Disease Management, Children's Hospital, Capital Medical University, National Centre for Children's Health, Beijing, China
| | - Caidie He
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Nian Wu
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Yingkuan Tian
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Songlin An
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Wei Chen
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Xiang Liu
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Haonan Zhang
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Shimin Xiong
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Yijun Liu
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Quan Li
- Department of Obstetrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuanzhong Zhou
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Xubo Shen
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| |
Collapse
|
34
|
Li S, Cao C, Watson D, Yang L, Kharbanda EO. Maternal smoking during pregnancy links to childhood blood pressure through birth weight and body mass index: NHANES 1999-2018. J Hum Hypertens 2024; 38:134-139. [PMID: 37773293 DOI: 10.1038/s41371-023-00865-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/22/2023] [Accepted: 06/21/2023] [Indexed: 10/01/2023]
Abstract
Maternal smoking during pregnancy (MSDP) is associated with lower birth weight, childhood obesity, and elevated blood pressure (BP) in offspring. We aimed to examine whether birth weight and body mass index (BMI) mediate the effect of MSDP on BP in children. The study included 14,713 children aged 8 to 15 years from the National Health and Nutrition Examination Surveys from 1999 to 2018. General third-variable models were used to examine the mediating effects of birth weight and BMI on the association of MSDP with BP. A total of 1928 (13.1%) children were exposed to MSDP. MSDP was associated with reduced birth weight (p < 0.001), increased BMI (p < 0.001), and elevated systolic BP (p = 0.005). MSDP was not associated with systolic BP after adjustment for birth weight and BMI z-score (p = 0.875), with 95.0% of the effect of MSDP on BP mediated by birth weight (39.1%) and BMI (55.9%). In conclusion, lower birth weight and increased obesity measures mediate the adverse effects of MSDP on BP in children. These findings provide novel mechanistic insight into the adverse effect of MSDP on BP in children and have implications for preventing hypertension in later life.
Collapse
Affiliation(s)
- Shengxu Li
- Children's Minnesota Research Institute, Children's Minnesota, Minneapolis, MN, USA.
| | - Chao Cao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dave Watson
- Children's Minnesota Research Institute, Children's Minnesota, Minneapolis, MN, USA
| | - Lin Yang
- Department of Cancer Epidemiology and Prevention Research, Cancer Research & Analytics, Cancer Care Alberta, Alberta Health Services, Calgary, AB, Canada
- Departments of Oncology and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
35
|
Maitin-Shepard M, Werner EF, Feig LA, Chavarro JE, Mumford SL, Wylie B, Rando OJ, Gaskins AJ, Sakkas D, Arora M, Kudesia R, Lujan ME, Braun J, Mozaffarian D. Food, nutrition, and fertility: from soil to fork. Am J Clin Nutr 2024; 119:578-589. [PMID: 38101699 DOI: 10.1016/j.ajcnut.2023.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Food and nutrition-related factors, including foods and nutrients consumed, dietary patterns, use of dietary supplements, adiposity, and exposure to food-related environmental contaminants, have the potential to impact semen quality and male and female fertility; obstetric, fetal, and birth outcomes; and the health of future generations, but gaps in evidence remain. On 9 November 2022, Tufts University's Friedman School of Nutrition Science and Policy and the school's Food and Nutrition Innovation Institute hosted a 1-d meeting to explore the evidence and evidence gaps regarding the relationships between food, nutrition, and fertility. Topics addressed included male fertility, female fertility and gestation, and intergenerational effects. This meeting report summarizes the presentations and deliberations from the meeting. Regarding male fertility, a positive association exists with a healthy dietary pattern, with high-quality evidence for semen quality and lower quality evidence for clinical outcomes. Folic acid and zinc supplementation have been found to not impact male fertility. In females, body weight status and other nutrition-related factors are linked to nearly half of all ovulation disorders, a leading cause of female infertility. Females with obesity have worse fertility treatment, pregnancy-related, and birth outcomes. Environmental contaminants found in food, water, or its packaging, including lead, perfluorinated alkyl substances, phthalates, and phenols, adversely impact female reproductive outcomes. Epigenetic research has found that maternal and paternal dietary-related factors can impact outcomes for future generations. Priority evidence gaps identified by meeting participants relate to the effects of nutrition and dietary patterns on fertility, gaps in communication regarding fertility optimization through changes in nutritional and environmental exposures, and interventions impacting germ cell mechanisms through dietary effects. Participants developed research proposals to address the priority evidence gaps. The workshop findings serve as a foundation for future prioritization of scientific research to address evidence gaps related to food, nutrition, and fertility.
Collapse
Affiliation(s)
| | - Erika F Werner
- Tufts University School of Medicine, Boston, MA, United States
| | - Larry A Feig
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA, United States
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Sunni L Mumford
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Blair Wylie
- Collaborative for Women's Environmental Health, Columbia University, New York, NY, United States
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Audrey J Gaskins
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, United States
| | | | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Marla E Lujan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Joseph Braun
- Department of Epidemiology, Brown University, Providence, RI, United States
| | - Dariush Mozaffarian
- Tufts University School of Medicine, Boston, MA, United States; Food is Medicine Institute, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States.
| |
Collapse
|
36
|
Bjørnsbo KS, Brøns C, Aadahl M, Kampmann FB, Friis Bryde Nielsen C, Lundbergh B, Wibaek R, Kårhus LL, Madsen AL, Hansen CS, Nørgaard K, Jørgensen NR, Suetta C, Kjaer M, Grarup N, Kanters J, Larsen M, Køber L, Kofoed KF, Loos R, Hansen T, Linneberg A, Vaag A. Protocol for the combined cardiometabolic deep phenotyping and registry-based 20-year follow-up study of the Inter99 cohort. BMJ Open 2024; 14:e078501. [PMID: 38286704 PMCID: PMC10826573 DOI: 10.1136/bmjopen-2023-078501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION The population-based Inter99 cohort has contributed extensively to our understanding of effects of a systematic screening and lifestyle intervention, as well as the multifactorial aetiology of type 2 diabetes (T2D) and cardiovascular disease. To understand causes, trajectories and patterns of early and overt cardiometabolic disease manifestations, we will perform a combined clinical deep phenotyping and registry follow-up study of the now 50-80 years old Inter99 participants. METHODS AND ANALYSIS The Inter99 cohort comprises individuals aged 30-60 years, who lived in a representative geographical area of greater Copenhagen, Denmark, in 1999. Age-stratified and sex-stratified random subgroups were invited to participate in either a lifestyle intervention (N=13 016) or questionnaires (N=5264), while the rest served as a reference population (N=43 021). Of the 13 016 individuals assigned to the lifestyle intervention group, 6784 (52%) accepted participation in a baseline health examination in 1999, including screening for cardiovascular risk factors and prediabetic conditions. In total, 6004 eligible participants, who participated in the baseline examination, will be invited to participate in the deep phenotyping 20-year follow-up clinical examination including measurements of anthropometry, blood pressure, arterial stiffness, cardiometabolic biomarkers, coronary artery calcification, heart rate variability, heart rhythm, liver stiffness, fundus characteristics, muscle strength and mass, as well as health and lifestyle questionnaires. In a subsample, 10-day monitoring of diet, physical activity and continuous glucose measurements will be performed. Fasting blood, urine and faecal samples to be stored in a biobank. The established database will form the basis of multiple analyses. A main purpose is to investigate whether low birth weight independent of genetics, lifestyle and glucose tolerance predicts later common T2D cardiometabolic comorbidities. ETHICS AND DISSEMINATION The study was approved by the Medical Ethics Committee, Capital Region, Denmark (H-20076231) and by the Danish Data Protection Agency through the Capital Region of Denmark's registration system (P-2020-1074). Informed consent will be obtained before examinations. Findings will be disseminated in peer-reviewed journals, at conferences and via presentations to stakeholders, including patients and public health policymakers. TRIAL REGISTRATION NUMBER NCT05166447.
Collapse
Affiliation(s)
- Kirsten Schroll Bjørnsbo
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | | - Mette Aadahl
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Freja Bach Kampmann
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Camilla Friis Bryde Nielsen
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Bjørn Lundbergh
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | | - Line Lund Kårhus
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Anja Lykke Madsen
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | | - Kirsten Nørgaard
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Charlotte Suetta
- Institute of Sports Medicine, Department of Orthopedic Surgery and Department of Geriatrics and Palliative Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine, Department of Orthopedic Surgery and Department of Geriatrics and Palliative Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Jørgen Kanters
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Center of Physiological Research, University of California, San Francisco, CA, USA
| | - Michael Larsen
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology and Radiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Klaus Fuglsang Kofoed
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology and Radiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ruth Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Lund University Diabetes Center, Malmö, Sweden
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
37
|
Saeed H, Wu J, Tesfaye M, Grantz KL, Tekola-Ayele F. Placental accelerated aging in antenatal depression. Am J Obstet Gynecol MFM 2024; 6:101237. [PMID: 38012987 PMCID: PMC10843762 DOI: 10.1016/j.ajogmf.2023.101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Antenatal maternal depression is associated with poor pregnancy outcomes and long-term effects on the offspring. Previous studies have identified links between antenatal depression and placental DNA methylation and between placental epigenetic aging and poor pregnancy outcomes, such as preterm labor and preeclampsia. The relationship between antenatal depression and poor pregnancy outcomes may be partly mediated via placental aging. OBJECTIVE This study aimed to investigate whether antenatal depressive symptoms are associated with placental epigenetic age acceleration, an epigenetic aging clock measure derived from the difference between methylation age and gestational age at delivery. STUDY DESIGN The study included 301 women who provided placenta samples at delivery as part of the Eunice Kennedy Shriver National Institute of Child Health and Human Development Fetal Growth Studies - Singletons that recruited participants from diverse race and ethnic groups at 12 US clinical sites (2009-2013). Women underwent depression screening using the Edinburgh Postnatal Depression Scale up to 6 times across the 3 trimesters of pregnancy. Depressive symptoms status was determined for each pregnancy trimester using an Edinburgh Postnatal Depression Scale score, in which a score of ≥10 was defined as having depressive symptoms and a score of <10 was defined as not having depressive symptoms. Placental DNA methylation was profiled from placenta samples. Placental epigenetic age was estimated using a methylation-based age estimator (placental "epigenetic clock") that has previously been found to have high placental gestational age prediction accuracy for uncomplicated term pregnancies. Placental age acceleration was defined to be the residual upon regressing the estimated epigenetic age on gestational age at delivery. Associations between an Edinburgh Postnatal Depression Scale score of ≥10 and an Edinburgh Postnatal Depression Scale score of <10 in the first, second, and third trimesters of pregnancy (ie, depressive symptoms vs none in each trimester) and placental age acceleration were tested using multivariable linear regression adjusting for maternal age, parity, race and ethnicity, and employment. RESULTS There were 31 (10.3%), 48 (16%), and 49 (16.4%) women with depressive symptoms (ie, Edinburgh Postnatal Depression Scale score of ≥10) in the first, second, and third trimesters of pregnancy, respectively. Of these women, 21 (7.2%) had sustained first- and second-trimester depressive symptoms, 19 (7%) had sustained second- and third-trimester depressive symptoms, and 12 (4.8%) had sustained depressive symptoms throughout pregnancy. Women with depressive symptoms in the second trimester of pregnancy had 0.41 weeks higher placental age acceleration than women without depressive symptoms during the second trimester of pregnancy (β=0.21 weeks [95% confidence interval, -0.17 to 0.58; P=.28] during the first trimester of pregnancy; β=0.41 weeks [95% confidence interval, 0.10-0.71; P=.009] during the second trimester of pregnancy; β=0.17 weeks [95% confidence interval, -0.14 to 0.47; P=.29] during the third trimester of pregnancy). Sustained first- and second-trimester depressive symptoms were associated with 0.72 weeks higher placental age acceleration (95% confidence interval, 0.29-1.15; P=.001) than no depressive symptom in the 2 trimesters. The association between second-trimester depressive symptoms and higher placental epigenetic age acceleration strengthened in the analysis of pregnancies with male fetuses (β=0.53 weeks; 95% confidence interval, 0.06-1.08; P=.03) but was not significant in pregnancies with female fetuses. CONCLUSION Antenatal depressive symptoms during the second trimester of pregnancy were associated with an average of 0.41 weeks of increased placental age acceleration. Accelerated placental aging may play an important role in the underlying mechanism linking antenatal depression to pregnancy complications related to placental dysfunction.
Collapse
Affiliation(s)
- Haleema Saeed
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD (Dr Saeed); Department of Maternal-Fetal Medicine, Medstar Washington Hospital Center, Washington, DC (Drs Saeed)
| | - Jing Wu
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Markos Tesfaye
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, University of Oslo, Oslo, Norway (Dr Tesfaye); Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Dr Tesfaye)
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele).
| |
Collapse
|
38
|
Al Mansoori A, Bataineh MF, Al Momani H, Ali HI. Micronutrient Status in Pregnant Women after Metabolic Bariatric Surgery in the United Arab Emirates: A Prospective Study. Nutrients 2023; 16:72. [PMID: 38201902 PMCID: PMC10781104 DOI: 10.3390/nu16010072] [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: 11/21/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Metabolic bariatric surgery (MBS) helps reduce comorbidities, such as hypertension and gestational diabetes, and is more effective than diet management for women with obesity-related health issues. Vitamin B12, vitamin D, and iron play important roles in ensuring the health of a neonate. However, pregnancies occurring after MBS may face complications related to micronutrient deficiencies, particularly of vitamins B12 and D and iron. This study aimed to investigate the vitamin B12, vitamin D, ferritin, and iron status of pregnant women who underwent MBS compared with women without MBS history. The study included 217 pregnant women (105 with a history of MBS and 112 without a history of MBS) who visited a major maternity hospital in Abu Dhabi, United Arab Emirates (UAE) between July 2021 and November 2022. The maternal vitamin B12, vitamin D, iron, and ferritin levels were measured twice, initially during the first or second trimester and subsequently during the third trimester. The iron was measured once during the pregnancy. Vitamin B12 deficiency was higher among pregnant women with MBS history compared to non-bariatric pregnant women (24.4% vs. 3.9%, p < 0.001). Women with a history of MBS had a higher prevalence of vitamin D deficiency (62.3% vs. 37.7%, p < 0.002). Linear regression analysis indicated that vitamin B12 levels decreased by 55 pg/mL in women with a history of MBS and by 4.6 pg/mL with a unit increase in body mass index (kg/m2). Furthermore, vitamin D levels in women with a history of MBS decreased by 4.9 ng/mL during pregnancy. Metabolic bariatric surgery is associated with vitamin B12, vitamin D, and iron deficiencies during pregnancy.
Collapse
Affiliation(s)
- Amna Al Mansoori
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.M.); (M.F.B.)
| | - Mo’ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.M.); (M.F.B.)
| | - Hazem Al Momani
- Weight Management Unit, NMC Royal Khalifa Hospital, Abu Dhabi P.O. Box 35233, United Arab Emirates;
| | - Habiba I. Ali
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.M.); (M.F.B.)
| |
Collapse
|
39
|
Cheddadi R, Yeramilli V, Martin C. From Mother to Infant, from Placenta to Gut: Understanding Varied Microbiome Profiles in Neonates. Metabolites 2023; 13:1184. [PMID: 38132866 PMCID: PMC10745069 DOI: 10.3390/metabo13121184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
The field of human microbiome and gut microbial diversity research has witnessed a profound transformation, driven by advances in omics technologies. These advancements have unveiled essential connections between microbiome alterations and severe conditions, prompting the development of new frameworks through epidemiological studies. Traditionally, it was believed that each individual harbored unique microbial communities acquired early in life, evolving over the course of their lifetime, with little acknowledgment of any prenatal microbial development, but recent research challenges this belief. The neonatal microbiome's onset, influenced by factors like delivery mode and maternal health, remains a subject of intense debate, hinting at potential intrauterine microbial processes. In-depth research reveals associations between microbiome profiles and specific health outcomes, ranging from obesity to neurodevelopmental disorders. Understanding these diverse microbiome profiles is essential for unraveling the intricate relationships between the microbiome and health outcomes.
Collapse
Affiliation(s)
- Riadh Cheddadi
- Department of Surgery, Division of Pediatric Surgery, Washington University School of Medicine, Saint Louis, MO 63110, USA (C.M.)
| | | | | |
Collapse
|
40
|
Cruz LL, Barco VS, Paula VG, Souza MR, Gallego FQ, Monteiro GC, Lima GPP, Damasceno DC, Volpato GT. Toxicological effects of the Curatella americana extract in embryo development of female pups from diabetic rats. Reprod Biol 2023; 23:100819. [PMID: 37918046 DOI: 10.1016/j.repbio.2023.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Maternal diabetes can influence the development of offspring during fetal life and postnatally. Curatella americana is a plant used as a menstrual cycle regulator and to prevent diabetes. This study evaluates the effects of C. americana aqueous extract on the estrous cycle and preimplantation embryos of adult female pups from diabetic rats. Female Sprague Dawley newborn rats received Streptozotocin or vehicle (citrate buffer). At adulthood, were submitted to the Oral Glucose Tolerance Test, and mated. The female rats were obtained and were distributed into four experimental groups: OC and OC/T represent female pups of control mothers and received water or plant extract, respectively; OD and OD/T represent female pups of diabetic mothers and received water or plant extract, respectively. The estrous cycle was followed for 10 days, the rats were mated and on gestational day 4 was performed preimplantation embryo analysis. Phenolic composition and biogenic amines in the extract were analyzed about the influence of the thermal process. The female pups from diabetic dams exhibited glucose intolerance, irregular estral cycle and a higher percentage of pre-embryos in delayed development (morula stage). After C. americana treatment, OD/T group no present a regular estrous cycle. Furthermore, the infusion process increases phenolic compounds and biogenic amines levels, which can have anti-estrogenic effect, anticipates the early embryonic development, and impair pre-implantation embryos. Thus, the indiscriminate use of medicinal plants should be avoided in any life phases by women, especially during pregnancy.
Collapse
Affiliation(s)
- Larissa Lopes Cruz
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Vinícius Soares Barco
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Maysa Rocha Souza
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Gean Charles Monteiro
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University, Botucatu, Brazil
| | - Giuseppina Pace Pereira Lima
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University, Botucatu, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Brazil.
| |
Collapse
|
41
|
Bao J, Gao Z, Hu Y, Ye L, Wang L. Transient receptor potential vanilloid type 1: cardioprotective effects in diabetic models. Channels (Austin) 2023; 17:2281743. [PMID: 37983306 PMCID: PMC10761101 DOI: 10.1080/19336950.2023.2281743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Cardiovascular disease, especially heart failure (HF) is the leading cause of death in patients with diabetes. Individuals with diabetes are prone to a special type of cardiomyopathy called diabetic cardiomyopathy (DCM), which cannot be explained by heart diseases such as hypertension or coronary artery disease, and can contribute to HF. Unfortunately, the current treatment strategy for diabetes-related cardiovascular complications is mainly to control blood glucose levels; nonetheless, the improvement of cardiac structure and function is not ideal. The transient receptor potential cation channel subfamily V member 1 (TRPV1), a nonselective cation channel, has been shown to be universally expressed in the cardiovascular system. Increasing evidence has shown that the activation of TRPV1 channel has a potential protective influence on the cardiovascular system. Numerous studies show that activating TRPV1 channels can improve the occurrence and progression of diabetes-related complications, including cardiomyopathy; however, the specific mechanisms and effects are unclear. In this review, we summarize that TRPV1 channel activation plays a protective role in the heart of diabetic models from oxidation/nitrification stress, mitochondrial function, endothelial function, inflammation, and cardiac energy metabolism to inhibit the occurrence and progression of DCM. Therefore, TRPV1 may become a latent target for the prevention and treatment of diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
42
|
Bailey SC, Pack AP, Wismer G, Calderon N, Velazquez E, Batio S, Ekong A, Eggleston A, Wallia A, Wolf MS, Schauer JM, Tenfelde S, Liebovitz DM, Grobman WA. Promoting REproductive Planning And REadiness in Diabetes (PREPARED) Study protocol: a clinic-randomised controlled trial testing a technology-based strategy to promote preconception care for women with type 2 diabetes. BMJ Open 2023; 13:e078282. [PMID: 37940161 PMCID: PMC10632823 DOI: 10.1136/bmjopen-2023-078282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023] Open
Abstract
INTRODUCTION Women with type 2 diabetes (T2DM) are more likely to experience adverse reproductive outcomes, yet preconception care can significantly reduce these risks. For women with T2DM, preconception care includes reproductive planning and patient education on: (1) the importance of achieving glycaemic control before pregnancy, (2) using effective contraception until pregnancy is desired, (3) discontinuing teratogenic medications if pregnancy could occur, (4) taking folic acid, and (5) managing cardiovascular and other risks. Despite its importance, few women with T2DM receive recommended preconception care. METHODS AND ANALYSIS We are conducting a two-arm, clinic-randomised trial at 51 primary care practices in Chicago, Illinois to evaluate a technology-based strategy to 'hardwire' preconception care for women of reproductive age with T2DM (the PREPARED (Promoting REproductive Planning And REadiness in Diabetes) strategy) versus usual care. PREPARED leverages electronic health record (EHR) technology before and during primary care visits to: (1) promote medication safety, (2) prompt preconception counselling and reproductive planning, and (3) deliver patient-friendly educational tools to reinforce counselling. Post-visit, text messaging is used to: (4) encourage healthy lifestyle behaviours. English and Spanish-speaking women, aged 18-44 years, with T2DM will be enrolled (N=840; n=420 per arm) and will receive either PREPARED or usual care based on their clinic's assignment. Data will be collected from patient interviews and the EHR. Outcomes include haemoglobin A1c (primary), reproductive knowledge and self-management behaviours. We will use generalised linear mixed-effects models (GLMMs) to evaluate the impact of PREPARED on these outcomes. GLMMs will include a fixed effect for treatment assignment (PREPARED vs usual care) and random clinic effects. ETHICS AND DISSEMINATION This study was approved by the Northwestern University Institutional Review Board (STU00214604). Study results will be published in journals with summaries shared online and with participants upon request. TRIAL REGISTRATION NUMBER ClinicalTrials.gov Registry (NCT04976881).
Collapse
Affiliation(s)
- Stacy C Bailey
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Allison P Pack
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Guisselle Wismer
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Norma Calderon
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Evelyn Velazquez
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stephanie Batio
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | - Amisha Wallia
- Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael S Wolf
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jacob M Schauer
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sandi Tenfelde
- Family and Community Health Department, Marcella Niehoff School of Nursing, Loyola University Chicago, Chicago, Illinois, USA
| | - David M Liebovitz
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - William A Grobman
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
43
|
Abstract
Diabetes in pregnancy affects 20 million women per year and is associated with increased risk of obesity in offspring, leading to insulin resistance and cardiometabolic disease. Despite the substantial public health ramifications, relatively little is known about the pathophysiological mechanisms underlying obesity in these high-risk children, which creates a barrier to successful intervention. While maternal glucose itself is undeniably a major stimulus upon intrauterine growth, the degree of offspring hyperinsulinism and disturbed lipid metabolism in mothers and offspring are also likely to be implicated in the disease process. The aim of this review is to summarise current understanding of the pathophysiology of childhood obesity after intrauterine exposure to maternal hyperglycaemia and to highlight possible opportunities for intervention. I present here a new unified hypothesis for the pathophysiology of childhood obesity in infants born to mothers with diabetes, which involves self-perpetuating twin cycles of pancreatic beta cell hyperfunction and altered lipid metabolism, both acutely and chronically upregulated by intrauterine exposure to maternal hyperglycaemia.
Collapse
Affiliation(s)
- Claire L Meek
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
44
|
Waage CW, Toftemo I, Brænd AM, Sletner L, Sommer C, Birkeland KI, Richardsen KR, Shakeel N, Vøllestad NK, Jenum AK. Cohort profile update: the Norwegian STORK Groruddalen (STORK G) pregnancy and birth cohort-the role of ethnicity and causal pathways for obesity, type 2 diabetes, cardiovascular disease and other health issues. BMJ Open 2023; 13:e076251. [PMID: 37899145 PMCID: PMC10619061 DOI: 10.1136/bmjopen-2023-076251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
PURPOSE The STORK Groruddalen cohort was set up in 2008 to explore ethnic differences in: (1) maternal health, primarily gestational diabetes (GDM) and related health issues during pregnancy and post partum, and effects of exposures on risk for type 2 diabetes, cardiovascular disease and other health issues, and (2) offspring's growth and body composition, overweight/obesity and effects of early life exposures. PARTICIPANTS 823 women (74% of invited) were followed from gestational week (GW) 15. Data were collected from 618 fathers. In total, 59% of women and 53% of fathers had origin from non-Western countries. Maternal mean age was 29.9 years (SD 4.9), and body mass index (BMI) 25.3 kg/m2 (4.9). Data were obtained from 772 women (94%) at GW 28, and 662 women (80%) 14 weeks post partum. Eleven years post partum, 385 women (53% of eligible/47% of original cohort) attended, age was 42.0 years (4.8) and BMI 27.1 kg/m2 (5.1). We have data for 783 children at birth, and for 586 at last time point, mean age 8.6 (0.5) years, weight 30.7 (6.8) kg and length 133.9 (6.3) cm. FINDINGS TO DATE We collected questionnaire data from parents, clinical measurements and blood samples from mothers, and data on children's growth (mid-pregnancy to 8 years). Our biobank includes maternal blood and urine samples, biopsy material from placentas and umbilical venous cord blood. We found several clinically important differences in maternal health, with higher risk in ethnic minority groups for GDM, insulin resistance, vitamin D and iron deficiency, depressive symptoms and physical inactivity. Contrasting patterns of fetal growth and risk of overweight/thinness at preschool age were observed across ethnic groups. Maternal GDM, obesity and high gestational weight gain were associated with children's BMI trajectories. FUTURE PLANS We will examine the impact of maternal and fetal health and development during pregnancy on long-term outcomes for mothers and offspring. TRIAL REGISTRATION NUMBER Project title STORK G-2: Women and Risk of Type 2 Diabetes NCT03870724 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Christin W Waage
- Department of General Practice, University of Oslo, Oslo, Norway
- Department of Rehabilitation Science and Health Technology, Oslo Metropolitan University, Oslo, Akershus, Norway
| | - Ingun Toftemo
- Department of General Practice, University of Oslo, Oslo, Norway
| | - Anja Maria Brænd
- Department of General Practice, University of Oslo, Oslo, Norway
- General Practice Research Unit (AFE), University of Oslo, Oslo, Norway
- The Antibiotic Centre for Primary Care, Department of General Practice, University of Oslo, Oslo, Norway
| | - Line Sletner
- Department of Child and Adolescents Medicine, Akershus University Hospital, Lorenskog, Norway
| | - Christine Sommer
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Kåre Inge Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Department of Transplantation, University of Oslo, Oslo, Norway
| | - Kåre Rønn Richardsen
- Department of Rehabilitation Science and Health Technology, Oslo Metropolitan University, Oslo, Akershus, Norway
| | - Nilam Shakeel
- Department of General Practice, University of Oslo, Oslo, Norway
| | - Nina Køpke Vøllestad
- Department of Interdisciplinary Health Science, University of Oslo, Oslo, Norway
| | - Anne Karen Jenum
- General Practice Research Unit (AFE), University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Kramer AC, Jansson T, Bale TL, Powell TL. Maternal-fetal cross-talk via the placenta: influence on offspring development and metabolism. Development 2023; 150:dev202088. [PMID: 37831056 PMCID: PMC10617615 DOI: 10.1242/dev.202088] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Compelling epidemiological and animal experimental data demonstrate that cardiometabolic and neuropsychiatric diseases originate in a suboptimal intrauterine environment. Here, we review evidence suggesting that altered placental function may, at least in part, mediate the link between the maternal environment and changes in fetal growth and development. Emerging evidence indicates that the placenta controls the development and function of several fetal tissues through nutrient sensing, modulation of trophoblast nutrient transporters and by altering the number and cargo of released extracellular vesicles. In this Review, we discuss the development and functions of the maternal-placental-fetal interface (in humans and mice) and how cross-talk between these compartments may be a mechanism for in utero programming, focusing on mechanistic target of rapamycin (mTOR), adiponectin and O-GlcNac transferase (OGT) signaling. We also discuss how maternal diet and stress influences fetal development and metabolism and how fetal growth restriction can result in susceptibility to developing chronic disease later in life. Finally, we speculate how interventions targeting placental function may offer unprecedented opportunities to prevent cardiometabolic disease in future generations.
Collapse
Affiliation(s)
- Avery C. Kramer
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Thomas Jansson
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Tracy L. Bale
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Theresa L. Powell
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
46
|
Gaggi G, Di Credico A, Barbagallo F, Ghinassi B, Di Baldassarre A. Bisphenols and perfluoroalkyls alter human stem cells integrity: A possible link with infertility. ENVIRONMENTAL RESEARCH 2023; 235:116487. [PMID: 37419196 DOI: 10.1016/j.envres.2023.116487] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023]
Abstract
Bisphenols and Perfluoroalkyls are chemical compounds widely used in industry known to be endocrine disruptors (EDs). Once ingested through contaminated aliments, they mimic the activity of endogenous hormones leading to a broad spectrum of diseases. Due to the extensive use of plastic in human life, particular attention should be paid to antenatal exposure to Bisphenols and Perfluoroalkyls since they cross the placental barrier and accumulates in developing embryo. Here we investigated the effects of Bisphenol-A (BPA), Bisphenol-S (BPS), perfluorooctane-sulfonate (PFOS) and perfluorooctanoic-acid (PFOA), alone or combined, on human-induced pluripotent stem cells (hiPSCs) that share several biological features with the stem cells of blastocysts. Our data show that these EDs affect hiPSC inducing a great mitotoxicity and dramatic changes in genes involved in the maintenance of pluripotency, germline specification, and epigenetic regulation. We also evidenced that these chemicals, when combined, may have additive, synergistic but also negative effects. All these data suggest that antenatal exposure to these EDs may affect the integrity of stem cells in the developing embryos, interfering with critical stages of early human development that might be determinant for fertility. The observation that the effects of exposure to a combination of these chemicals are not easily foreseeable further highlights the need for wider awareness of the complexity of the EDs effects on human health and of the social and economic burden attributable to these compounds.
Collapse
Affiliation(s)
- Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | - Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy.
| | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| |
Collapse
|
47
|
Abstract
Eating behaviours are determined by the integration of interoceptive and environmental inputs. During pregnancy, numerous physiological adaptations take place in the maternal organism to provide an adequate environment for embryonic growth. Among them, whole-body physiological remodelling directly influences eating patterns, commonly causing notable taste perception alterations, food aversions and cravings. Recurrent food cravings for and compulsive eating of highly palatable food can contribute to the development and maintenance of gestational overweight and obesity with potential adverse health consequences for the offspring. Although much is known about how maternal eating habits influence offspring health, the mechanisms that underlie changes in taste perception and food preference during pregnancy (which guide and promote feeding) are only just starting to be elucidated. Given the limited and diffuse understanding of the neurobiology of gestational eating patterns, the aim of this Review is to compile, integrate and discuss the research conducted on this topic in both experimental models and humans. This article sheds light on the mechanisms that drive changes in female feeding behaviours during distinct physiological states. Understanding these processes is crucial to improve gestational parent health and decrease the burden of metabolic and food-related diseases in future generations.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
48
|
Spencer R, Maksym K, Hecher K, Maršál K, Figueras F, Ambler G, Whitwell H, Nené NR, Sebire NJ, Hansson SR, Diemert A, Brodszki J, Gratacós E, Ginsberg Y, Weissbach T, Peebles DM, Zachary I, Marlow N, Huertas-Ceballos A, David AL. Maternal PlGF and umbilical Dopplers predict pregnancy outcomes at diagnosis of early-onset fetal growth restriction. J Clin Invest 2023; 133:e169199. [PMID: 37712421 PMCID: PMC10503803 DOI: 10.1172/jci169199] [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: 02/01/2023] [Accepted: 06/27/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUNDSevere, early-onset fetal growth restriction (FGR) causes significant fetal and neonatal mortality and morbidity. Predicting the outcome of affected pregnancies at the time of diagnosis is difficult, thus preventing accurate patient counseling. We investigated the use of maternal serum protein and ultrasound measurements at diagnosis to predict fetal or neonatal death and 3 secondary outcomes: fetal death or delivery at or before 28+0 weeks, development of abnormal umbilical artery (UmA) Doppler velocimetry, and slow fetal growth.METHODSWomen with singleton pregnancies (n = 142, estimated fetal weights [EFWs] below the third centile, less than 600 g, 20+0 to 26+6 weeks of gestation, no known chromosomal, genetic, or major structural abnormalities) were recruited from 4 European centers. Maternal serum from the discovery set (n = 63) was analyzed for 7 proteins linked to angiogenesis, 90 additional proteins associated with cardiovascular disease, and 5 proteins identified through pooled liquid chromatography and tandem mass spectrometry. Patient and clinician stakeholder priorities were used to select models tested in the validation set (n = 60), with final models calculated from combined data.RESULTSThe most discriminative model for fetal or neonatal death included the EFW z score (Hadlock 3 formula/Marsal chart), gestational age, and UmA Doppler category (AUC, 0.91; 95% CI, 0.86-0.97) but was less well calibrated than the model containing only the EFW z score (Hadlock 3/Marsal). The most discriminative model for fetal death or delivery at or before 28+0 weeks included maternal serum placental growth factor (PlGF) concentration and UmA Doppler category (AUC, 0.89; 95% CI, 0.83-0.94).CONCLUSIONUltrasound measurements and maternal serum PlGF concentration at diagnosis of severe, early-onset FGR predicted pregnancy outcomes of importance to patients and clinicians.TRIAL REGISTRATIONClinicalTrials.gov NCT02097667.FUNDINGThe European Union, Rosetrees Trust, Mitchell Charitable Trust.
Collapse
Affiliation(s)
- Rebecca Spencer
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Kasia Maksym
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karel Maršál
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences Lund, Skane University Hospital, Lund University, Lund, Sweden
| | - Francesc Figueras
- Institut D’Investigacions Biomèdiques August Pi í Sunyer, University of Barcelona, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Barcelona, Spain
| | - Gareth Ambler
- Department of Statistical Science, University College London, London, United Kingdom
| | - Harry Whitwell
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction and
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nuno Rocha Nené
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
| | - Neil J. Sebire
- Population, Policy and Practice Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Stefan R. Hansson
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences Lund, Skane University Hospital, Lund University, Lund, Sweden
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Brodszki
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences Lund, Skane University Hospital, Lund University, Lund, Sweden
| | - Eduard Gratacós
- Institut D’Investigacions Biomèdiques August Pi í Sunyer, University of Barcelona, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Barcelona, Spain
| | - Yuval Ginsberg
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
- Department of Obstetrics and Gynecology, Rambam Medical Centre, Haifa, Israel
| | - Tal Weissbach
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
- Department of Obstetrics and Gynecology, Sheba Medical Center Tel Hashomer, Tel Aviv, Israel
| | - Donald M. Peebles
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
| | - Ian Zachary
- Division of Medicine, Faculty of Medical Sciences, University College London, United Kingdom
| | - Neil Marlow
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
| | - Angela Huertas-Ceballos
- Neonatal Department, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Anna L. David
- UCL Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, United Kingdom
| |
Collapse
|
49
|
Xie Q, Zhang X, Zhou Q, Xu Y, Sun L, Wen Q, Wang W, Chen Q. Antioxidant and anti-inflammatory properties of ginsenoside Rg1 for hyperglycemia in type 2 diabetes mellitus: systematic reviews and meta-analyses of animal studies. Front Pharmacol 2023; 14:1179705. [PMID: 37745069 PMCID: PMC10514510 DOI: 10.3389/fphar.2023.1179705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/02/2023] [Indexed: 09/26/2023] Open
Abstract
Background: According to existing laboratory data, ginsenoside Rg1 may help cure diabetes and its complications by reducing oxidative stress (OS) and managing inflammation. However, this conclusion lacks reliability and is unclear. As a result, the purpose of this systematic review and meta-analysis was to evaluate the antioxidant and anti-inflammatory effects of ginsenoside Rg1 in the treatment of diabetes and its complications. Methods: We searched for relevant studies published through December 2022, including electronic bibliographic databases such as PubMed, EMBASE, Web of Science, CNKI, and Wanfang. The SYstematic Review Center for Laboratory Animal Experimentation Risk of Bias (SYRCLE RoB) tool was used to conduct a meta-analysis to assess the methodological quality of animal research. The meta-analysis was conducted using RevMan5.4 software, following the Cochrane Handbook for Systematic Reviews of Interventions. This study is registered in the International Systems Review Prospective Registry (PROSPERO) as CRD42023386830. Results: Eighteen eligible studies involving 401 animals were included. Ginsenoside Rg1 was significantly correlated with blood glucose (BG), insulin levels, body weight, superoxide dismutase (SOD), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) levels. In addition, according to subgroup analysis, the hypoglycemic, anti-inflammatory, and antioxidant effects of ginsenoside Rg1 in type 2 diabetic animals were not affected by experimental species, modeling, experimental drug dosage, or course of treatment. Conclusion: This meta-analysis presents a summary of the hypoglycemic effects of ginsenoside Rg1, which are achieved through anti-inflammatory and antioxidant mechanisms. These findings provide evidence-based support for the medical efficacy of ginsenoside Rg1. Specifically, ginsenoside Rg1 reduced MDA levels and restored SOD activity to exert its antioxidant activity. It had a positive effect on the reduction of IL-6 and TNF-α levels. However, the inclusion of studies with low methodological quality and the presence of publication bias may undermine the validity of the results. Further investigation with a more rigorous experimental design and comprehensive studies is necessary to fully understand the specific glycemic mechanisms of ginsenosides. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier https://CRD42023386830.
Collapse
Affiliation(s)
- Qian Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoran Zhang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumei Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lisha Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Wen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Wang
- School of Biomedical Sciences, Mianyang Normal University, Mianyang, China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
50
|
Lyssenko V, Vaag A. Genetics of diabetes-associated microvascular complications. Diabetologia 2023; 66:1601-1613. [PMID: 37452207 PMCID: PMC10390394 DOI: 10.1007/s00125-023-05964-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/27/2023] [Indexed: 07/18/2023]
Abstract
Diabetes is associated with excess morbidity and mortality due to both micro- and macrovascular complications, as well as a range of non-classical comorbidities. Diabetes-associated microvascular complications are those considered most closely related to hyperglycaemia in a causal manner. However, some individuals with hyperglycaemia (even those with severe hyperglycaemia) do not develop microvascular diseases, which, together with evidence of co-occurrence of microvascular diseases in families, suggests a role for genetics. While genome-wide association studies (GWASs) produced firm evidence of multiple genetic variants underlying differential susceptibility to type 1 and type 2 diabetes, genetic determinants of microvascular complications are mostly suggestive. Identified susceptibility variants of diabetic kidney disease (DKD) in type 2 diabetes mirror variants underlying chronic kidney disease (CKD) in individuals without diabetes. As for retinopathy and neuropathy, reported risk variants currently lack large-scale replication. The reported associations between type 2 diabetes risk variants and microvascular complications may be explained by hyperglycaemia. More extensive phenotyping, along with adjustments for unmeasured confounding, including both early (fetal) and late-life (hyperglycaemia, hypertension, etc.) environmental factors, are urgently needed to understand the genetics of microvascular complications. Finally, genetic variants associated with reduced glycolysis, mitochondrial dysfunction and DNA damage and sustained cell regeneration may protect against microvascular complications, illustrating the utility of studies in individuals who have escaped these complications.
Collapse
Affiliation(s)
- Valeriya Lyssenko
- Department of Clinical Science, Mohn Research Center for Diabetes Precision Medicine, University of Bergen, Bergen, Norway.
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Lund, Sweden.
| | - Allan Vaag
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Lund, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|