1
|
Laporta J, Dado-Senn B, Guadagnin AR, Liu L, Peñagaricano F. Preweaning heat stress alters liver transcriptome and DNA methylation in dairy calves. J Dairy Sci 2025; 108:4390-4402. [PMID: 39892602 DOI: 10.3168/jds.2024-25975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/31/2024] [Indexed: 02/04/2025]
Abstract
Prenatal hyperthermia has long-lasting effects on dairy calf metabolism, immunity, and productivity. Yet, the effects of postnatal heat stress on neonatal calves remain unclear. As such, this study aimed to investigate the impact of heat stress on the preweaning dairy calf liver gene expression and DNA methylation profiles. Holstein dairy calves were exposed to summer heat stress (daily average temperature-humidity index >78) for 42 d postnatally (i.e., birth to weaning) with access to active fan heat abatement (postnatal cooling, post-CL; calf-height air speed 2.05 m/s, n = 12) or not (postnatal heat stress, post-HS; calf-height air speed 0.15 m/s, n = 12). All thermoregulatory responses were lower in post-CL relative to post-HS calves throughout the experimental period (-12 bpm, -2.5°C, and -0.11°C for respiratory frequency, rectal temperature, and ear skin temperature, respectively). Liver tissue was harvested via biopsy (n = 6 male calves per treatment) at 42 d of age for RNA sequencing and reduced representation bisulfite sequencing. There were 73 differentially expressed genes (DEG), of which 31 were downregulated and 42 were upregulated in post-HS relative to post-CL treatments (false discovery rate cut-off, 20%). Ingenuity pathways analysis revealed that post-HS significantly affected 24 pathways and 60 transcription regulators, including pathway PI3K/AKT, and transcription regulators PPARGC1A, STAT5B, CREB, and XBP1. A total of 14,639 differentially methylated cytosines (DMC) were found across the entire bovine genome; these DMC mapped to 3,197 differentially methylated genes (DMG), with about 300 DMG with DMC located close to the transcription start site. These DMG, such as PKA, AMPK, MAPK, and STAT3, are closely related to metabolic signaling pathways. Overall, preweaning exposure of dairy calves to heat stress changes hepatic methylation profiles, which in turn may affect the expression of genes with critical roles in intracellular signaling and development, metabolic, and immune-related pathways. Providing mechanical cooling via fans to dairy calves in summer seems beneficial to promoting thermoregulation and liver cellular hemostasis.
Collapse
Affiliation(s)
- Jimena Laporta
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706.
| | - Bethany Dado-Senn
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Anne R Guadagnin
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Lihe Liu
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Francisco Peñagaricano
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
2
|
Liu S, Xu L, Cheng Y, Liu D, Zhang B, Chen X, Zheng M. Methylation of the telomerase gene promoter region in umbilical cord blood of patients with gestational diabetes mellitus is associated with decreased telomerase expression levels and shortened telomere length. Front Endocrinol (Lausanne) 2025; 16:1502329. [PMID: 40134806 PMCID: PMC11932890 DOI: 10.3389/fendo.2025.1502329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Objective This study speculates that gestational diabetes mellitus (GDM) may reduce fetal telomere length (TL),which may be related to modification of methylation in the promoter region of the telomerase (TE) gene promoter region. Methods In this study, umbilical cord blood samples from patients with and without GDM (N = 100 each) were analyzed by prospective case-control. The TL, TE expression levels, and methylation levels of TERT and TERC gene promoter regions in two groups were measured. The significance of the methylation level of each CpG locus employed logistic regression analysis of R software, and the analysis of covariance (ANCOVA) was used to control the influence of confounding factors. Correlation analysis was performed by the Spearman. Results The TL and TE expression levels of the offspring of GDM patients were decreased despite adjusting for PBMI, PWG, and TG. A total of two CpG islands were screened in the promoter region of the TERT gene and three fragments (TERT_2, TERT_3, and TERT_4) containing a total of 70 CpG sites were designed. Additionally, four CpG sites of the TERT gene in the GDM group (TERT_2_40, TERT_2_47, TERT_3_46, and TERT_3_212) showed increased methylation levels compared with the control group (all P < 0.05). In the promoter region of the TERC gene, one CpG island containing 19 CpG loci was screened and designed, and the methylation levels of the two CpG sites were significantly different in TERC_1_67 (0.65 ± 0.21 versus 0.57 ± 0.30; P = 0.040) and TERC_1_120 (0.68 ± 0.23 versus 0.59 ± 0.27; P = 0.014). The methylation levels of TERC gene fragments of GDM patients were significantly higher than those of the control group (0.69 ± 0.06 versus 0.65 ± 0.08, P = 0.001). Conclusion This study revealed that GDM may induce decreased TE expression by increasing the methylation levels of TE genes promoter region, thereby reducing the TL.
Collapse
Affiliation(s)
- Shuhua Liu
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Liping Xu
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yan Cheng
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Dehong Liu
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Bin Zhang
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Xianxia Chen
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Mingming Zheng
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Mattera MSDLC, Scaramele NF, Lopes FL, Belardi BE, Tsosura TVS, Sampaio HM, Chiba FY, Pereira RF, Dos Santos RM, Ervolino E, Baliero GF, Nobumoto ACTY, Cachoni AC, Chaves-Neto AH, Matsushita DH. MicroRNA expression profiling in the adult offspring of rats with periodontal disease. Arch Oral Biol 2025; 170:106131. [PMID: 39566203 DOI: 10.1016/j.archoralbio.2024.106131] [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: 07/02/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE The present study investigated the relationship between maternal periodontal disease, insulin resistance, activation of inflammatory pathways and epigenetic modifications in adult offspring. DESIGN Therefore, female Wistar rats were divided into control and experimental groups. Seven days after the induction of periodontal disease, female rats from both groups were mated with healthy male rats. After weaning, male offspring were divided into control offspring (CN-o) and periodontal disease offspring (PED-o) groups. Body weight was measured at 0-75 days of age. At day 75, the following were measured in the offspring: insulin resistance by the HOMA-IR index; global miRNAs by microtranscriptome array; validation of the selected miRNAs by quantitative real-time PCR expression; interleukin 1 receptor associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6) content in the gastrocnemius muscle tissue (GSM) by western blotting. RESULTS Maternal periodontal disease leads to low birth weight (LBW) in the offspring and insulin resistance in adulthood; changes in global miRNA expression (5 miRNAs upregulated and 6 downregulated); and increased protein expression of IRAK1 and TRAF6 in GSM. CONCLUSIONS These findings demonstrate that maternal periodontal disease causes LBW, insulin resistance, activation of inflammatory pathways, and changes in global miRNA expression.
Collapse
Affiliation(s)
- Maria Sara de Lima Coutinho Mattera
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil.
| | | | - Flávia Lombardi Lopes
- São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Bianca Elvira Belardi
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Thaís Verônica Saori Tsosura
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Heloisa Macedo Sampaio
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Fernando Yamamoto Chiba
- Department of Preventive and Restorative Dentistry, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Renato Felipe Pereira
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Rodrigo Martins Dos Santos
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Gabriele Fernandes Baliero
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Ana Carla Thalez Ywabuchi Nobumoto
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Anna Clara Cachoni
- Department of Preventive and Restorative Dentistry, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Antonio Hernandes Chaves-Neto
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Doris Hissako Matsushita
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil; Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| |
Collapse
|
4
|
Nguyen HT, Tran HTT, Dao-Tran TH, Huang LC. Nutrition literacy in Vietnamese pregnant women: a cross-sectional study. Health Promot Int 2025; 40:daae187. [PMID: 39820456 DOI: 10.1093/heapro/daae187] [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] [Indexed: 01/19/2025] Open
Abstract
Inadequate nutrition intake during pregnancy elevates the risk of adverse health outcomes during pregnancy, with potential long-term repercussions for both mother and child, extending to subsequent generations. Current initiatives to improve individual dietary habits emphasize promoting nutrition literacy (NL), which encompasses the ability to access, comprehend, and use basic nutrition information and services necessary for making appropriate nutrition decisions. However, there were limited data on the NL of pregnant women in Vietnam. Therefore, this study aims to explore the NL levels of Vietnamese pregnant women and examine the factors related to their NL. A total of 360 Vietnamese pregnant women participated in the study from May to September 2023. A validated questionnaire (Nutrition Literacy Assessment Instrument for Pregnant Women, NLAI-P), assessing the knowledge, behavior and skill, was applied. A general linear model with univariate linear regression analysis was conducted to identify predictor factors of NL. The findings revealed that 70.3% of participants had inadequate NL. Among the three dimensions, nutrition knowledge was particularly low, with no respondents achieving an adequate level and 94.7% scoring at an inadequate level. High household monthly income, age, normal prepregnancy weight and indoor work were statistically associated with higher NL scores. This study highlights the limited NL among Vietnamese pregnant women. Increasing NL is crucial for supporting their optimal healthy diet, enhancing the health of pregnant women and their offspring and future generations.
Collapse
Affiliation(s)
- Hoan Thi Nguyen
- Department of Health Care Science, China Medical University, 100, Sec. 1, Jingmao Road, Taichung 406040, Taiwan
- Faculty of Nursing and Medical Technology, University of Medicine and Pharmacy at Ho Chi Minh City, 201 Nguyen Chi Thanh, District 5, Ho Chi Minh City, Vietnam
| | - Hang Thi Thuy Tran
- University of Medicine and Pharmacy at Ho Chi Minh City Hospital, 216 Hong Bang, District 5, Ho Chi Minh City 17000, Vietnam
| | - Tiet-Hanh Dao-Tran
- Center of Health Services Research, Faculty of Medicine, University of Queensland, Level 5, UQ Health Science Building, Herston Campus, Brisban, QLD4006,Australia
| | - Li-Chi Huang
- Department of Health Care Science, China Medical University, 100, Sec. 1, Jingmao Road, Taichung 406040, Taiwan
- School of Nursing, China Medical University, 100, Jingmao Road, Beitun District, Taichung 406040, Taiwan
- Department of Nursing, China Medical University Children Hospital, 100, Sec. 1, Jingmao Road, Taichung 406040, Taiwan
| |
Collapse
|
5
|
Wang S, Peng R, Chen H, Han D, Wu J, Xu Y, Ying Z. Timing determines programming of energy homeostasis by maternal PM 2.5 exposure in mouse models. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125187. [PMID: 39454811 PMCID: PMC11602340 DOI: 10.1016/j.envpol.2024.125187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Maternal exposure to particulate matter with an aerodynamic diameter ≤2.5 μm (PM2.5) is believed to be a risk factor of developmental origins of health and disease (DOHaD), but its effect on offspring's susceptibility to obesity, a common target disease of DOHaD, remains controversial. To pinpoint the effect of maternal PM2.5 exposure on offspring's energy homeostasis, female C57BL/6J mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) for 12 weeks and mated with normal male mice to produce offspring. After parturition, a cross-fostering strategy was exploited to determine whether prenatal and/or postnatal mothering by CAP-exposed dams program offspring's energy homeostasis and susceptibility to obesity. Moreover, oocytes were collected from FA- or CAP-exposed mice and subjected to in vitro fertilization (IVF) to determine whether maternal pre-conceptional exposure to PM2.5 programs energy homeostasis. Results showed that prenatal mothering by CAP-exposed dams increased suckling's milk intake and weight gain, decreased normal diet (ND)-fed offspring's adulthood food intake and body weight, and did not influence offspring's diet-induced obesity (DIO). Postnatal mothering by CAP-exposed dams did not influence suckling's milk intake and weight gain, increased ND-fed offspring's adulthood food intake and body weight and did not influence offspring's DIO. Prenatal plus postnatal mothering by CAP-exposed dams increased suckling's milk intake and weight gain, increased ND-fed offspring's adulthood food intake and body weight, and aggravated offspring's DIO. IVF study revealed that male offspring derived from CAP-exposed mice versus controls had significantly decreased adulthood food intake and body weight. RNA sequencing showed that CAP exposure influenced oocyte estrogen signaling and histone methylation. This study thus clearly reveals that timing determines programming of energy homeostasis by maternal PM2.5 exposure.
Collapse
Affiliation(s)
- Shan Wang
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Renzhen Peng
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Hongxia Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
6
|
Bozack AK, Trasande L. Prenatal chemical exposures and the methylome: current evidence and opportunities for environmental epigenetics. Epigenomics 2024; 16:1443-1451. [PMID: 39539208 PMCID: PMC11622816 DOI: 10.1080/17501911.2024.2426441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Exposure to pollutants and chemicals during critical developmental periods in early life can impact health and disease risk across the life course. Research in environmental epigenetics has provided increasing evidence that prenatal exposures affect epigenetic markers, particularly DNA methylation. In this article, we discuss the role of DNA methylation in early life programming and review evidence linking the intrauterine environment to epigenetic modifications, with a focus on exposure to tobacco smoke, metals, and endocrine-disrupting chemicals. We also discuss challenges and novel approaches in environmental epigenetic research and explore the potential of epigenetic biomarkers in studies of pediatric populations as indicators of exposure and disease risk. Overall, we aim to highlight how advancements in environmental epigenetics may transform our understanding of early-life exposures and inform new approaches for supporting long-term health.
Collapse
Affiliation(s)
- Anne K. Bozack
- Department of Epidemiology and Population Health, Stanford School of Medicine, Palo Alto, CA, USA
| | - Leonardo Trasande
- Department of Pediatrics and Department of Population Health, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
7
|
Huo M, Yu X, Yuan X, Guo J, Wei B, Shi Y, Gu Y, Zhang X, Sun M. The P300-ARRDC3 axis participates in maternal subclinical hypothyroidism and is involved in abnormal hepatic insulin sensitivity in adult offspring. Heliyon 2024; 10:e39259. [PMID: 39568856 PMCID: PMC11577204 DOI: 10.1016/j.heliyon.2024.e39259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Numerous studies have suggested potential associations between maternal subclinical hypothyroidism (SCH) and adverse metabolic outcomes in offspring, however, the underlying mechanism remains unclear. In this study, we generated a maternal SCH mouse model by administering 50 ppm 6-propyl-2-thiouracil (PTU) in the drinking water of pregnant mice until delivery. This model was used to investigate the mechanisms influencing glucose metabolism in offspring. RNA sequencing (RNA-seq) revealed a substantial increase in ARRDC3 expression in the livers of the offspring of the SCH model mice, which may contribute to insulin resistance. Additionally, the phosphorylation levels of key proteins in the insulin signalling pathway, such as protein kinase B (Akt), glycogen synthase kinase 3 beta (GSK-3β), and Forkhead box protein O1 (FoxO1), were correspondingly reduced in the SCH offspring. Moreover, overexpression of ARRDC3 in Hepa1‒6 cells suppressed the Akt/GSK-3β/FoxO1 signalling pathway and increased the expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), which was consistent with the molecular changes observed in SCH offspring. Our results also indicated that the upregulation of ARRDC3 in SCH offspring may result from increased H3K27 acetylation of the ARRDC3 promoter region, driven by elevated expression of P300. Importantly, adequate L-T4 supplementation during pregnancy improved insulin sensitivity and reversed the molecular alterations in the insulin signalling pathway observed in SCH offspring. In conclusion, exposure to intrauterine SCH resulted in altering the P300-ARRDC3 axis in offspring and impaired insulin sensitivity by disrupting the Akt/GSK-3β/FoxO1 signalling pathway. Timely L-T4 supplementation during pregnancy is an effective strategy to prevent insulin resistance in offspring of SCH mothers. This study elucidates potential molecular mechanisms behind insulin resistance in SCH offspring and suggests novel therapeutic targets for treating metabolic disorders related to maternal SCH.
Collapse
Affiliation(s)
- Ming Huo
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou City, 730000, Gansu, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xianbin Yuan
- The first people's hospital of Lanzhou City, Lanzhou City, 730000, Gansu, China
| | - Jun Guo
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yannan Gu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xuehong Zhang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou City, 730000, Gansu, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
8
|
Mukoyama M. Fetal origins of adult hypertension and renal injury: an epigenetic memory matter? Hypertens Res 2024; 47:2942-2944. [PMID: 39138365 DOI: 10.1038/s41440-024-01812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Masashi Mukoyama
- Department of Nephrology, Omuta Tenryo Hospital, Omuta, Fukuoka, Japan.
| |
Collapse
|
9
|
Muraglia M, Faienza MF, Tardugno R, Clodoveo ML, Matias De la Cruz C, Bermúdez FG, Munizaga MG, Valencia L, Corbo F, Orellana-Manzano A. Breastfeeding: science and knowledge in pediatric obesity prevention. Front Med (Lausanne) 2024; 11:1430395. [PMID: 39399112 PMCID: PMC11466875 DOI: 10.3389/fmed.2024.1430395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
The increasing prevalence of childhood obesity worldwide is a significant concern due to its link to severe health issues in adulthood, such as non-communicable diseases (NCDs). To address this issue, this review evaluates the effectiveness of various preventive measures for childhood obesity, focusing on maternal nutrition and breastfeeding. The study underscores the criticality of the periconceptional period, where the diets of both parents can influence epigenetic modifications that impact the child's metabolic pathways and obesity risks. Breastfeeding is a potent protective mechanism against early-onset obesity, significantly enhancing the infant's metabolic and immune health by modifying DNA methylation and gene expression. Furthermore, the perspective underscores the significance of the Mediterranean diet during the periconceptional period and lactation. This diet can effectively prevent gestational complications and improve breast milk quality, fostering optimal infant development. Recognizing that obesity results from genetic, epigenetic, environmental, and social factors, the paper advocates for a comprehensive, multidisciplinary approach from the earliest stages of life. This approach champions a balanced maternal diet, exclusive breastfeeding, and timely introduction to complementary foods. In conclusion, addressing pediatric obesity requires a multifaceted strategy emphasizing improving prenatal and postnatal nutrition. Further research is necessary to understand the epigenetic mechanisms influenced by nutrition and their long-term effects on children's health. This will help refine interventions that curb the obesity epidemic among future generations.
Collapse
Affiliation(s)
- Marilena Muraglia
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, Bari, Italy
| | - Roberta Tardugno
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Lisa Clodoveo
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari-Aldo Moro, Bari, Italy
| | - Carmen Matias De la Cruz
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Fátima German Bermúdez
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - María Gabriela Munizaga
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Luz Valencia
- Licenciatura en Nutrición y Dietética, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Filomena Corbo
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Andrea Orellana-Manzano
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| |
Collapse
|
10
|
Snyder LR, Koutmou KS. Studying the intersection of nucleoside modifications and SARS-CoV-2 RNA-dependent RNA transcription using an in vitro reconstituted system. Methods Enzymol 2024; 705:81-109. [PMID: 39389674 PMCID: PMC11849750 DOI: 10.1016/bs.mie.2024.07.005] [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] [Indexed: 10/12/2024]
Abstract
There is growing recognition that viral RNA genomes possess enzymatically incorporated modified nucleosides. These small chemical changes are analogous to epigenomic modifications in DNA and have the potential to be similarly important modulators of viral transcription and evolution. However, the molecular level consequences of individual sites of modification remain to be broadly explored. Here we describe an in vitro assay to examine the impact of nucleoside modifications on the rate and fidelity of SARS-CoV-2 RNA transcription. Establishing the role of modified nucleotides in SARS-CoV-2 is of interest both for advancing fundamental knowledge of RNA modifications in viruses, and because modulating the modification-landscape of SARS-CoV-2 may represent a therapeutic strategy to interfere with viral RNA replication. Our approach can be used to assess the influence both of modifications present in a template RNA, as well nucleotide analog inhibitors. These methods provide a reproducible guide for generating active SARS-CoV-2 replication/transcription complexes capable of establishing how RNA modifications influence the pre-steady state rate constants of nucleotide addition by RNA-dependent RNA polymerases.
Collapse
Affiliation(s)
- Laura R Snyder
- Department of Chemistry, University of Michigan, Ann Arbor, MI, United States
| | - Kristin S Koutmou
- Department of Chemistry, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
11
|
Adegboyega O, Gayen Nee' Betal S, Urday P, Huang R, Bodycot K, Al-Kouatly HB, Solarin K, Chan JSY, Addya S, Boelig RC, Aghai ZH. DNA methylation patterns in umbilical cord blood from infants of methadone maintained opioid dependent mothers. Sci Rep 2024; 14:17298. [PMID: 39068260 PMCID: PMC11283475 DOI: 10.1038/s41598-024-66899-w] [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: 03/17/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024] Open
Abstract
Methadone maintenance treatment for opioid dependent mothers is standard of care. Infants of methadone maintained opioid dependent (MMOD) mothers have better outcomes compared to infants of opioid dependent mothers without treatment. However, when compared to non-exposed infants, infants of MMOD mothers are associated with worse outcomes. We conducted a pilot study to examine genome wide differential DNA methylation using cord blood samples from sixteen term and near-term infants of MMOD and opioid naïve mothers, excluding Infants with chorioamnionitis. A total of 152 differentially methylated loci were identified at a difference > + 2, < - 2 and p-value < 0.05. There were 90 hypermethylated loci (59 annotated genes) and 62 hypomethylated loci (38 annotated genes) observed. The hypermethylated and hypomethylated DNA changes involved multiple genes, pathways and networks that may explain some of the changes seen in infants of MMOD mothers. Top hypermethylated and hypomethylated genes involved areas of cell growth, neurodevelopment, vision and xenobiotic metabolism functions. Our data may explain the role of key pathways and genes relevant to neonatal outcomes seen from methadone exposure in pregnancy. Functional studies on the identified pathways and genes could lead to improved understanding of the mechanisms and identify areas for intervention.
Collapse
Affiliation(s)
- Oluwatobi Adegboyega
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
- Reilly Children's Hospital, Lehigh Valley Health Network, Allentown, USA
| | - Suhita Gayen Nee' Betal
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Pedro Urday
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Rachel Huang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Katherine Bodycot
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Huda B Al-Kouatly
- Division of Maternal Fetal Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Kolawole Solarin
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Joanna S Y Chan
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Sankar Addya
- Laboratory of Cancer Genomics, Thomas Jefferson University, Philadelphia, USA
| | - Rupsa C Boelig
- Division of Maternal Fetal Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Zubair H Aghai
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Basak S, Mallick R, Navya Sree B, Duttaroy AK. Placental Epigenome Impacts Fetal Development: Effects of Maternal Nutrients and Gut Microbiota. Nutrients 2024; 16:1860. [PMID: 38931215 PMCID: PMC11206482 DOI: 10.3390/nu16121860] [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: 05/02/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish an exposure threshold due to nutrient deficiencies or other environmental factors that can result in clinically relevant epigenetic alterations that modulate disease risks in the fetus. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development. This review unravels the potential roles of the functional epigenome for targeted intervention to ensure optimal fetal brain development and its performance in later life.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Boga Navya Sree
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
13
|
Yoshida A, Takata Y, Tabara Y, Maruyama K, Inoue S, Osawa H, Sugiyama T. Interaction effect between low birthweight and resistin gene rs1862513 variant on insulin resistance and type 2 diabetes mellitus in adulthood: Toon Genome Study. J Diabetes Investig 2024; 15:725-735. [PMID: 38421160 PMCID: PMC11143422 DOI: 10.1111/jdi.14163] [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: 10/31/2023] [Revised: 01/11/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
AIMS/INTRODUCTION Gene-environment interactions are considered to critically influence type 2 diabetes mellitus development; however, the underlying mechanisms and specific interactions remain unclear. Given the increasing prevalence of low birthweight (LBW) influenced by the intrauterine environment, we sought to investigate genetic factors related to type 2 diabetes development in individuals with LBW. MATERIALS AND METHODS The interaction between 20 reported type 2 diabetes susceptibility genes and the development of type 2 diabetes in LBW (<2,500 g) individuals in a population-based Japanese cohort (n = 1,021) was examined by logistic regression and stratified analyses. RESULTS Logistic regression analyses showed that only the G/G genotype at the rs1862513 locus of the resistin gene (RETN), an established initiator of insulin resistance, was closely related to the prevalence of type 2 diabetes in individuals with LBW. Age, sex and current body mass index-adjusted stratified analyses showed a significant interaction effect of LBW and the RETN G/G genotype on fasting insulin, homeostatic model assessment 2-insulin resistance, Matsuda index and the prevalence of type 2 diabetes (all P-values for interaction <0.05). The adjusted odds ratio for type 2 diabetes in the LBW + G/G genotype group was 7.33 (95% confidence interval 2.43-22.11; P = 0.002) compared with the non-LBW + non-G/G genotype group. Similar results were obtained after excluding the influence of malnutrition due to World War II. CONCLUSIONS Simultaneous assessment of LBW and the RETN G/G genotype can more accurately predict the risk of future type 2 diabetes than assessing each of these factors alone, and provide management strategies, including early lifestyle intervention in LBW population.
Collapse
Affiliation(s)
- Ayaka Yoshida
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| | - Yasunori Takata
- Department of Diabetes and Molecular GeneticsEhime University Graduate School of MedicineToonEhimeJapan
| | - Yasuharu Tabara
- Center for Genomic MedicineKyoto University Graduate School of MedicineKyotoJapan
- Graduate School of Public HealthShizuoka Graduate University of Public HealthShizuokaJapan
| | - Koutatsu Maruyama
- Department of Bioscience, Graduate School of AgricultureEhime UniversityToonEhimeJapan
| | - Shota Inoue
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| | - Haruhiko Osawa
- Department of Diabetes and Molecular GeneticsEhime University Graduate School of MedicineToonEhimeJapan
| | - Takashi Sugiyama
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| |
Collapse
|
14
|
Sámano R, Martínez-Rojano H, Chico-Barba G, Gamboa R, Tolentino M, Toledo-Barrera AX, Ramírez-González C, Mendoza-Flores ME, Hernández-Trejo M, Godínez-Martínez E. Serum Folate, Red Blood Cell Folate, and Zinc Serum Levels Are Related with Gestational Weight Gain and Offspring's Birth-Weight of Adolescent Mothers. Nutrients 2024; 16:1632. [PMID: 38892565 PMCID: PMC11174574 DOI: 10.3390/nu16111632] [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] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Gestational weight gain below or above the Institute of Medicine recommendations has been associated with adverse perinatal and neonatal outcomes. Very few studies have evaluated the association between serum and red blood cell folate concentrations and gestational weight gain in adolescents. Additionally, zinc deficiency during pregnancy has been associated with impaired immunity, prolonged labor, preterm and post-term birth, intrauterine growth restriction, low birth weight, and pregnancy-induced hypertension. OBJECTIVE The purpose of our study is to evaluate the association between serum concentrations of zinc, serum folate, and red blood cell folate, with the increase in gestational weight and the weight and length of the newborn in a group of adolescent mothers from Mexico City. RESULTS In our study, 406 adolescent-neonate dyads participated. The adolescents' median age was 15.8 years old. The predominant socioeconomic level was middle-low (57.8%), single (57%), 89.9% were engaged in home activities, and 41.3% completed secondary education. Excessive gestational weight gain was observed in 36.7% of cases, while insufficient gestational weight gain was noted in 38.4%. Small for gestational age infants were observed in 20.9% of the sample. Low serum folate (OR 2.1, 95% CI 1.3-3.3), decreased red blood cell folate (OR 1.6, 95% CI 1.0-2.6), and reduced serum zinc concentrations (OR 3.3, 95% CI 2.1-5.2) were associated with insufficient gestational weight gain. Decreased serum zinc levels (OR 1.2, 95% CI 1.2-3.4) were linked to an increased probability of delivering a baby who is small for their gestational age. CONCLUSIONS Low serum folate, red blood cell folate, and serum zinc concentrations were associated with gestational weight gain and having a small gestational age baby. Both excessive and insufficient gestational weight gain, as well as having a small gestational age baby, are frequent among adolescent mothers.
Collapse
Affiliation(s)
- Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| | - Hugo Martínez-Rojano
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, México City 11340, Mexico
- Coordinación de Medicina Laboral, Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) “Dr. Manuel Martínez Báez”, Secretaría de Salud, México City 01480, Mexico
| | - Gabriela Chico-Barba
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| | - Ricardo Gamboa
- Departamento de Fisiología, Instituto Nacional de Cardiología, México City 14080, Mexico;
| | - Maricruz Tolentino
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| | | | - Cristina Ramírez-González
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| | - María Eugenia Mendoza-Flores
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| | - María Hernández-Trejo
- Departamento de Neurobiología del Desarrollo, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico;
| | - Estela Godínez-Martínez
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, México City 11000, Mexico; (G.C.-B.); (M.T.); (C.R.-G.); (M.E.M.-F.); (E.G.-M.)
| |
Collapse
|
15
|
Bakoyan Z, Cao Y, Hansson SR, Karlsson JP, Lodefalk M. Childhood atopic disorders in relation to placental changes-A systematic review and meta-analysis. Pediatr Allergy Immunol 2024; 35:e14141. [PMID: 38773752 DOI: 10.1111/pai.14141] [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: 10/12/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024]
Abstract
Fetal programming may arise from prenatal exposure and increase the risk of diseases later in life, potentially mediated by the placenta. The objective of this systematic review was to summarize and critically evaluate publications describing associations between human placental changes and risk of atopic disorders during childhood. The review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. The inclusion criteria were original research articles or case reports written in English describing a human placental change in relation to disease occurring in offspring during childhood. The MEDLINE and EMBASE databases were searched for eligible studies. Risk of bias (RoB) was assessed using the ROBINS-I tool. The results were pooled both in a narrative way and by a meta-analysis. Nineteen studies were included (n = 12,997 participants). All studies had an overall serious RoB, and publication bias could not be completely ruled out. However, five studies showed that histological chorioamnionitis in preterm-born children was associated with asthma-related problems (pooled odds ratio = 3.25 (95% confidence interval = 2.22-4.75)). In term-born children, a large placenta (≥750 g) increased the risk of being prescribed anti-asthma medications during the first year of life. Placental histone acetylation, DNA methylation, and gene expression differences were found to be associated with different atopic disorders in term-born children. There is some evidence supporting the idea that the placenta can mediate an increased risk of atopic disorders in children. However, further studies are needed to validate the findings, properly control for confounders, and examine potential mechanisms.
Collapse
Affiliation(s)
- Zaki Bakoyan
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Science Lund, Lund University, Lund, Sweden
| | | | - Maria Lodefalk
- University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Pediatrics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
16
|
Smith SM, Bais B, Ismaili M'hamdi H, Schermer MH, Steegers-Theunissen RP. Stimulating Preconception Care Uptake by Women With a Vulnerable Health Status Through a Mobile Health App (Pregnant Faster): Pilot Feasibility Study. JMIR Hum Factors 2024; 11:e53614. [PMID: 38648092 DOI: 10.2196/53614] [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: 10/12/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND A low socioeconomic status is associated with a vulnerable health status (VHS) through the accumulation of health-related risk factors, such as poor lifestyle behaviors (eg, inadequate nutrition, chronic stress, and impaired health literacy). For pregnant women, a VHS translates into a high incidence of adverse pregnancy outcomes and therefore pregnancy-related inequity. We hypothesize that stimulating adequate pregnancy preparation, targeting lifestyle behaviors and preconception care (PCC) uptake, can reduce these inequities and improve the pregnancy outcomes of women with a VHS. A nudge is a behavioral intervention aimed at making healthy choices easier and more attractive and may therefore be a feasible way to stimulate engagement in pregnancy preparation and PCC uptake, especially in women with a VHS. To support adequate pregnancy preparation, we designed a mobile health (mHealth) app, Pregnant Faster, that fits the preferences of women with a VHS and uses nudging to encourage PCC consultation visits and engagement in education on healthy lifestyle behaviors. OBJECTIVE This study aimed to test the feasibility of Pregnant Faster by determining usability and user satisfaction, the number of visited PCC consultations, and the course of practical study conduction. METHODS Women aged 18-45 years, with low-to-intermediate educational attainment, who were trying to become pregnant within 12 months were included in this open cohort. Recruitment took place through social media, health care professionals, and distribution of flyers and posters from September 2021 until June 2022. Participants used Pregnant Faster daily for 4 weeks, earning coins by reading blogs on pregnancy preparation, filling out a daily questionnaire on healthy lifestyle choices, and registering for a PCC consultation with a midwife. Earned coins could be spent on rewards, such as fruit, mascara, and baby products. Evaluation took place through the mHealth App Usability Questionnaire (MAUQ), an additional interview or questionnaire, and assessment of overall study conduction. RESULTS Due to limited inclusions, the inclusion criterion "living in a deprived neighborhood" was dropped. This resulted in the inclusion of 47 women, of whom 39 (83%) completed the intervention. In total, 16 (41%) of 39 participants visited a PCC consultation, with their main motivation being obtaining personalized information. The majority of participants agreed with 16 (88.9%) of 18 statements of the MAUQ, indicating high user satisfaction. The mean rating was 7.7 (SD 1.0) out of 10. Points of improvement included recruitment of the target group, simplification of the log-in system, and automation of manual tasks. CONCLUSIONS Nudging women through Pregnant Faster to stimulate pregnancy preparation and PCC uptake has proven feasible, but the inclusion criteria must be revised. A substantial number of PCC consultations were conducted, and this study will therefore be continued with an open cohort of 400 women, aiming to establish the (cost-)effectiveness of an updated version, named Pregnant Faster 2. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR2-10.2196/45293.
Collapse
Affiliation(s)
- Sharissa M Smith
- Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Babette Bais
- Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hafez Ismaili M'hamdi
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Maartje Hn Schermer
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | |
Collapse
|
17
|
Luo J, Wang W, Li J, Duan H, Xu C, Tian X, Zhang D. Epigenome-wide association study identifies DNA methylation loci associated with handgrip strength in Chinese monozygotic twins. Front Cell Dev Biol 2024; 12:1378680. [PMID: 38633108 PMCID: PMC11021642 DOI: 10.3389/fcell.2024.1378680] [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: 01/30/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Background: The decline in muscle strength and function with aging is well recognized, but remains poorly characterized at the molecular level. Here, we report the epigenetic relationship between genome-wide DNA methylation and handgrip strength (HGS) among Chinese monozygotic (MZ) twins. Methods: DNA methylation (DNAm) profiling was conducted in whole blood samples through Reduced Representation Bisulfite Sequencing method. Generalized estimating equation was applied to regress the DNAm of each CpG with HGS. The Genomic Regions Enrichment of Annotations Tool was used to perform enrichment analysis. Differentially methylated regions (DMRs) were detected using comb-p. Causal inference was performed using Inference about Causation through Examination of Familial Confounding method. Finally, we validated candidate CpGs in community residents. Results: We identified 25 CpGs reaching genome-wide significance level. These CpGs located in 9 genes, especially FBLN1, RXRA, and ABHD14B. Many enriched terms highlighted calcium channels, neuromuscular junctions, and skeletal muscle organ development. We identified 21 DMRs of HGS, with several DMRs within FBLN1, SLC30A8, CST3, and SOCS3. Causal inference indicated that the DNAm of 16 top CpGs within FBLN1, RXRA, ABHD14B, MFSD6, and TYW1B might influence HGS, while HGS influenced DNAm at two CpGs within FBLN1 and RXRA. In validation analysis, methylation levels of six CpGs mapped to FLBN1 and one CpG mapped to ABHD14B were negatively associated with HGS weakness in community population. Conclusion: Our study identified multiple DNAm variants potentially related to HGS, especially CpGs within FBLN1 and ABHD14B. These findings provide new clues to the epigenetic modification underlying muscle strength decline.
Collapse
Affiliation(s)
- Jia Luo
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, Shandong, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, Shandong, China
| | - Jingxian Li
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, Shandong, China
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, Shandong, China
- Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, Shandong, China
- Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, Shandong, China
- Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
18
|
Prince S. Gene-environment interaction: why genetic enhancement might never be distributed fairly. JOURNAL OF MEDICAL ETHICS 2024; 50:272-277. [PMID: 37268408 DOI: 10.1136/jme-2023-109101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/19/2023] [Indexed: 06/04/2023]
Abstract
Ethical debates around genetic enhancement tend to include an argument that the technology will eventually be fairly accessible once available. That we can fairly distribute genetic enhancement has become a moral defence of genetic enhancement. Two distribution solutions are argued for, the first being equal distribution. Equality of access is generally believed to be the fairest and most just method of distribution. Second, equitable distribution: providing genetic enhancements to reduce social inequalities. In this paper, I make two claims. I first argue that the very assumption that genetic enhancements can be distributed fairly is problematic when considering our understanding of gene-environment interactions, for example, epigenetics. I then argue that arguments that genetic enhancements are permissible because the intended benefits can be distributed fairly as intended are misinformed. My first claim rests on the assertion that genetic enhancements do not enhance traits in a vacuum; genes are dependent on conducive environments for expression. If society cannot guarantee fair environments, then any benefit conferred from being genetically enhanced will be undermined. Thus, any argument that the distribution of genetic enhancements will be fair and that the technology is therefore morally permissible, is mistaken.
Collapse
Affiliation(s)
- Sinead Prince
- Faculty of Business and Law, Queensland University of Technology, Brisbane, Queensland, Australia
- Faculty of Business and Law, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
19
|
Sze Y, Brunton PJ. How is prenatal stress transmitted from the mother to the fetus? J Exp Biol 2024; 227:jeb246073. [PMID: 38449331 DOI: 10.1242/jeb.246073] [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] [Indexed: 03/08/2024]
Abstract
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo-pituitary-adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
- Zhejiang University-University of Edinburgh Joint Institute, Haining, Zhejiang 314400, P.R. China
| |
Collapse
|
20
|
Coker SJ, Berry MJ, Vissers MCM, Dyson RM. Maternal Vitamin C Intake during Pregnancy Influences Long-Term Offspring Growth with Timing- and Sex-Specific Effects in Guinea Pigs. Nutrients 2024; 16:369. [PMID: 38337653 PMCID: PMC10857109 DOI: 10.3390/nu16030369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Our previous work in guinea pigs revealed that low vitamin C intake during preconception and pregnancy adversely affects fertility, pregnancy outcomes, and foetal and neonatal growth in a sex-dependent manner. To investigate the long-term impact on offspring, we monitored their growth from birth to adolescence (four months), recorded organ weights at childhood equivalence (28 days) and adolescence, and assessed physiological parameters like oral glucose tolerance and basal cortisol concentrations. We also investigated the effects of the timing of maternal vitamin C restriction (early vs. late gestation) on pregnancy outcomes and the health consequences for offspring. Dunkin Hartley guinea pigs were fed an optimal (900 mg/kg feed) or low (100 mg/kg feed) vitamin C diet ad libitum during preconception. Pregnant dams were then randomised into four feeding regimens: consistently optimal, consistently low, low during early pregnancy, or low during late pregnancy. We found that low maternal vitamin C intake during early pregnancy accelerated foetal and neonatal growth in female offspring and altered glucose homeostasis in the offspring of both sexes at an age equivalent to early childhood. Conversely, low maternal vitamin C intake during late pregnancy resulted in foetal growth restriction and reduced weight gain in male offspring throughout their lifespan. We conclude that altered vitamin C during development has long-lasting, sex-specific consequences for offspring and that the timing of vitamin C depletion is also critical, with low levels during early development being associated with the development of a metabolic syndrome-related phenotype, while later deprivation appears to be linked to a growth-faltering phenotype.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| | - Mary J. Berry
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| | - Margreet C. M. Vissers
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Rebecca M. Dyson
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| |
Collapse
|
21
|
Ow MC, Hall SE. Inheritance of Stress Responses via Small Non-Coding RNAs in Invertebrates and Mammals. EPIGENOMES 2023; 8:1. [PMID: 38534792 DOI: 10.3390/epigenomes8010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 03/28/2024] Open
Abstract
While reports on the generational inheritance of a parental response to stress have been widely reported in animals, the molecular mechanisms behind this phenomenon have only recently emerged. The booming interest in epigenetic inheritance has been facilitated in part by the discovery that small non-coding RNAs are one of its principal conduits. Discovered 30 years ago in the Caenorhabditis elegans nematode, these small molecules have since cemented their critical roles in regulating virtually all aspects of eukaryotic development. Here, we provide an overview on the current understanding of epigenetic inheritance in animals, including mice and C. elegans, as it pertains to stresses such as temperature, nutritional, and pathogenic encounters. We focus on C. elegans to address the mechanistic complexity of how small RNAs target their cohort mRNAs to effect gene expression and how they govern the propagation or termination of generational perdurance in epigenetic inheritance. Presently, while a great amount has been learned regarding the heritability of gene expression states, many more questions remain unanswered and warrant further investigation.
Collapse
Affiliation(s)
- Maria C Ow
- Department of Biology, Syracuse University, Syracuse, NY 13210, USA
| | - Sarah E Hall
- Department of Biology and Program in Neuroscience, Syracuse University, Syracuse, NY 13210, USA
| |
Collapse
|
22
|
Meulders B, Marei WFA, Xhonneux I, Bols PEJ, Leroy JLMR. Effect of lipotoxicity on mitochondrial function and epigenetic programming during bovine in vitro embryo production. Sci Rep 2023; 13:21664. [PMID: 38066095 PMCID: PMC10709407 DOI: 10.1038/s41598-023-49184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
Maternal metabolic disorders may cause lipotoxic effects on the developing oocyte. Understanding the timing at which this might disrupt embryo epigenetic programming and how this is linked with mitochondrial dysfunction is crucial for improving assisted reproductive treatments, but has not been investigated before. Therefore, we used a bovine in vitro model to investigate if pathophysiological palmitic acid (PA) concentrations during in vitro oocyte maturation and in vitro embryo culture alter embryo epigenetic patterns (DNA methylation (5mC) and histone acetylation/methylation (H3K9ac/H3K9me2)) compared to control (CONT) and solvent control (SCONT), at the zygote and morula stage. Secondly, we investigated if these epigenetic alterations are associated with mitochondrial dysfunction and changes in ATP production rate, or altered expression of epigenetic regulatory genes. Compared to SCONT, H3K9ac and H3K9me2 levels were increased in PA-derived zygotes. Also, 5mC and H3K9me2 levels were increased in PA-exposed morulae compared to SCONT. This was associated with complete inhibition of glycolytic ATP production in oocytes, increased mitochondrial membrane potential and complete inhibition of glycolytic ATP production in 4-cell embryos and reduced SOD2 expression in PA-exposed zygotes and morulae. For the first time, epigenetic alterations in metabolically compromised zygotes and morulae have been observed in parallel with mitochondrial dysfunction in the same study.
Collapse
Affiliation(s)
- Ben Meulders
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium.
| | - Waleed F A Marei
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Inne Xhonneux
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Peter E J Bols
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Jo L M R Leroy
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
23
|
Ciancio A, Behrman J, Kämpfen F, Kohler IV, Maurer J, Mwapasa V, Kohler HP. Barker's Hypothesis Among the Global Poor: Positive Long-Term Cardiovascular Effects of in Utero Famine Exposure. Demography 2023; 60:1747-1766. [PMID: 37937904 PMCID: PMC10875974 DOI: 10.1215/00703370-11052790] [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] [Indexed: 11/09/2023]
Abstract
An influential literature on the Developmental Origins of Health and Disease (DOHaD) has documented that poor conditions in utero lead to higher risk of cardiovascular disease at older ages. Evidence from low-income countries (LICs) has hitherto been missing, despite the fact that adverse in utero conditions are far more common in LICs. We find that Malawians exposed in utero to the 1949 Nyasaland famine have better cardiovascular health 70 years later. These findings highlight the potential context specificity of the DOHaD hypothesis, with in utero adversity having different health implications among aging LIC individuals who were exposed to persistent poverty.
Collapse
Affiliation(s)
- Alberto Ciancio
- Adam Smith Business School, University of Glasgow, Glasgow, UK
| | - Jere Behrman
- Department of Economics, University of Pennsylvania, Philadelphia, PA, USA
| | - Fabrice Kämpfen
- School of Economics, University College Dublin, Dublin, Ireland
| | - Iliana V Kohler
- Population Studies Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jürgen Maurer
- Department of Economics, University of Lausanne, Lausanne, Switzerland
| | | | - Hans-Peter Kohler
- Population Studies Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Bueno López C, Gómez Moreno G, Palloni A. Empirical evidence of predictive adaptive response in humans: systematic review and meta-analysis of migrant populations. J Dev Orig Health Dis 2023; 14:728-745. [PMID: 38196328 PMCID: PMC12013326 DOI: 10.1017/s2040174423000429] [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] [Indexed: 01/11/2024]
Abstract
Meta-analysis is used to test a variant of a Developmental Origins of Adult Health and Disease (DOHaD)'s conjecture known as predictive adaptive response (PAR). According to it, individuals who are exposed to mismatches between adverse or constrained in utero conditions, on the one hand, and postnatal obesogenic environments, on the other, are at higher risk of developing adult chronic conditions, including obesity, type 2 diabetes (T2D), hypertension and cardiovascular disease. We argue that migrant populations from low and middle to high-income countries offer a unique opportunity to test the conjecture. A database was constructed from an exhaustive literature search of peer-reviewed papers published prior to May 2021 contained in PUBMED and SCOPUS using keywords related to migrants, DOHaD, and associated health outcomes. Random effects meta-regression models were estimated to assess the magnitude of effects associated with migrant groups on the prevalence rate of T2D and hypertension in adults and overweight/obesity in adults and children. Overall, we used 38 distinct studies and 78 estimates of diabetes, 59 estimates of hypertension, 102 estimates of overweight/obesity in adults, and 23 estimates of overweight/obesity in children. Our results show that adult migrants experience higher prevalence of T2D than populations at destination (PR 1.48; 95% CI 1.35-1.65) and origin (PR 1.80; 95% CI 1.40-2.34). Similarly, there is a significant excess of obesity prevalence in children migrants (PR 1.22; 95% CI 1.04-1.43) but not among adult migrants (PR 0.89; 95% CI 0.80-1.01). Although the total effect of migrant status on prevalence of hypertension is centered on zero, some migrant groups show increased risks. Finally, the size of estimated effects varies significantly by migrant groups according to place of destination. Despite limitations inherent to all meta-analyses and admitting that some of our findings may be accounted for alternative explanations, the present study shows empirical evidence consistent with selected PAR-like conjectures.
Collapse
Affiliation(s)
- Clara Bueno López
- Department of Population, Institute of Economy, Geography and Demography, Spanish National Research Council, Madrid, Spain
| | - Guillermo Gómez Moreno
- Department of Population, Institute of Economy, Geography and Demography, Spanish National Research Council, Madrid, Spain
| | - Alberto Palloni
- Department of Population, Institute of Economy, Geography and Demography, Spanish National Research Council, Madrid, Spain
- Center for Demography of Health and Aging (CDHA), University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
25
|
Kumar S, Seem K, Kumar S, Singh A, Krishnan SG, Mohapatra T. DNA methylome analysis provides insights into gene regulatory mechanism for better performance of rice under fluctuating environmental conditions: epigenomics of adaptive plasticity. PLANTA 2023; 259:4. [PMID: 37993704 DOI: 10.1007/s00425-023-04272-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
MAIN CONCLUSION Roots play an important role in adaptive plasticity of rice under dry/direct-sown conditions. However, hypomethylation of genes in leaves (resulting in up-regulated expression) complements the adaptive plasticity of Nagina-22 under DSR conditions. Rice is generally cultivated by transplanting which requires plenty of water for irrigation. Such a practice makes rice cultivation a challenging task under global climate change and reducing water availability. However, dry-seeded/direct-sown rice (DSR) has emerged as a resource-saving alternative to transplanted rice (TPR). Though some of the well-adapted local cultivars are used for DSR, only limited success has been achieved in developing DSR varieties mainly because of a limited knowledge of adaptability of rice under fluctuating environmental conditions. Based on better morpho-physiological and agronomic performance of Nagina-22 (N-22) under DSR conditions, N-22 and IR-64 were grown by transplanting and direct-sowing and used for whole genome methylome analysis to unravel the epigenetic basis of adaptive plasticity of rice. Comparative methylome and transcriptome analyses indicated a large number (4078) of genes regulated through DNA methylation/demethylation in N-22 under DSR conditions. Gene × environment interactions play important roles in adaptive plasticity of rice under direct-sown conditions. While genes for pectinesterase, LRK10, C2H2 zinc-finger protein, splicing factor, transposable elements, and some of the unannotated proteins were hypermethylated, the genes for regulation of transcription, protein phosphorylation, etc. were hypomethylated in CG context in the root of N-22, which played important roles in providing adaptive plasticity to N-22 under DSR conditions. Hypomethylation leading to up-regulation of gene expression in the leaf complements the adaptive plasticity of N-22 under DSR conditions. Moreover, differential post-translational modification of proteins and chromatin assembly/disassembly through DNA methylation in CHG context modulate adaptive plasticity of N-22. These findings would help developing DSR cultivars for increased water-productivity and ecological efficiency.
Collapse
Affiliation(s)
- Suresh Kumar
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India.
| | - Karishma Seem
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | | | - Archana Singh
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - S Gopala Krishnan
- Division of Genetics, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | | |
Collapse
|
26
|
Onuzulu CD, Lee S, Basu S, Comte J, Hai Y, Hizon N, Chadha S, Fauni MS, Kahnamoui S, Xiang B, Halayko AJ, Dolinsky VW, Pascoe CD, Jones MJ. Early-life exposure to cigarette smoke primes lung function and DNA methylation changes at Cyp1a1 upon exposure later in life. Am J Physiol Lung Cell Mol Physiol 2023; 325:L552-L567. [PMID: 37642652 PMCID: PMC11068412 DOI: 10.1152/ajplung.00192.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Prenatal and early-life exposure to cigarette smoke (CS) has repeatedly been shown to induce stable, long-term changes in DNA methylation (DNAm) in offspring. It has been hypothesized that these changes might be functionally related to the known outcomes of prenatal and early-life CS exposure, which include impaired lung development, altered lung function, and increased risk of asthma and wheeze. However, to date, few studies have examined DNAm changes induced by prenatal CS in tissues of the lung, and even fewer have attempted to examine the specific influences of prenatal versus early postnatal exposures. Here, we have established a mouse model of CS exposure which isolates the effects of prenatal and early postnatal CS exposures in early life. We have used this model to measure the effects of prenatal and/or postnatal CS exposures on lung function and immune cell infiltration as well as DNAm and expression of Cyp1a1, a candidate gene previously observed to demonstrate DNAm differences on CS exposure in humans. Our study revealed that exposure to CS prenatally and in the early postnatal period causes long-lasting differences in offspring lung function, gene expression, and lung Cyp1a1 DNAm, which wane over time but are reestablished on reexposure to CS in adulthood. This study creates a testable mouse model that can be used to investigate the effects of prenatal and early postnatal CS exposures and will contribute to the design of intervention strategies to mediate these detrimental effects.NEW & NOTEWORTHY Here, we isolated effects of prenatal from early postnatal cigarette smoke and showed that exposure to cigarette smoke early in life causes changes in offspring DNA methylation at Cyp1a1 that last through early adulthood but not into late adulthood. We also showed that smoking in adulthood reestablished these DNA methylation patterns at Cyp1a1, suggesting that a mechanism other than DNA methylation results in long-term memory associated with early-life cigarette smoke exposures at this gene.
Collapse
Affiliation(s)
- Chinonye Doris Onuzulu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Samantha Lee
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jeannette Comte
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Yan Hai
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nikho Hizon
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shivam Chadha
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maria Shenna Fauni
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shana Kahnamoui
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bo Xiang
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Vernon W Dolinsky
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christopher D Pascoe
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
27
|
Vitikainen EIK, Meniri M, Marshall HH, Thompson FJ, Businge R, Mwanguhya F, Kyabulima S, Mwesige K, Ahabonya S, Sanderson JL, Kalema-Zikusoka G, Hoffman JI, Wells D, Lewis G, Walker SL, Nichols HJ, Blount JD, Cant MA. The social formation of fitness: lifetime consequences of prenatal nutrition and postnatal care in a wild mammal population. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220309. [PMID: 37381858 PMCID: PMC10291432 DOI: 10.1098/rstb.2022.0309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 06/30/2023] Open
Abstract
Research in medicine and evolutionary biology suggests that the sequencing of parental investment has a crucial impact on offspring life history and health. Here, we take advantage of the synchronous birth system of wild banded mongooses to test experimentally the lifetime consequences to offspring of receiving extra investment prenatally versus postnatally. We provided extra food to half of the breeding females in each group during pregnancy, leaving the other half as matched controls. This manipulation resulted in two categories of experimental offspring in synchronously born litters: (i) 'prenatal boost' offspring whose mothers had been fed during pregnancy, and (ii) 'postnatal boost' offspring whose mothers were not fed during pregnancy but who received extra alloparental care in the postnatal period. Prenatal boost offspring lived substantially longer as adults, but postnatal boost offspring had higher lifetime reproductive success (LRS) and higher glucocorticoid levels across the lifespan. Both types of experimental offspring had higher LRS than offspring from unmanipulated litters. We found no difference between the two experimental categories of offspring in adult weight, age at first reproduction, oxidative stress or telomere lengths. These findings are rare experimental evidence that prenatal and postnatal investments have distinct effects in moulding individual life history and fitness in wild mammals. This article is part of the theme issue 'Evolutionary ecology of inequality'.
Collapse
Affiliation(s)
- E. I. K. Vitikainen
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
- Organismal and Evolutionary Biology, University of Helsinki, Helsinki, PO Box 65, 00014 Finland
| | - M. Meniri
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
| | - H. H. Marshall
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
- Centre for Research in Ecology, Evolution and Behaviour, University of Roehampton, Roehampton Lane, London SW15 5PJ, UK
| | - F. J. Thompson
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
| | - R. Businge
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
| | - F. Mwanguhya
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
| | - S. Kyabulima
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
| | - K. Mwesige
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
| | - S. Ahabonya
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
| | - J. L. Sanderson
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
| | - G. Kalema-Zikusoka
- Conservation Through Public Health, PO Box 75298, Uringi Crescent Rd, Entebbe, Uganda
| | - J. I. Hoffman
- Department of Behavioural Ecology, University of Bielefeld, Bielefeld, Konsequenz 45, 33619, Germany
| | - D. Wells
- Department of Behavioural Ecology, University of Bielefeld, Bielefeld, Konsequenz 45, 33619, Germany
| | - G. Lewis
- Department of Biosciences, Wallace Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - S. L. Walker
- Chester Zoo Endocrine Laboratory, Endocrinology, Science Centre, Caughall Road, Upton-by-Chester, Chester, CH2 1LH, UK
| | - H. J. Nichols
- Department of Biosciences, Wallace Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - J. D. Blount
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
| | - M. A. Cant
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
- Banded Mongoose Research Project, Queen Elizabeth National Park, PO Box 66 Lake Katwe, Kasese District, Uganda
- German Primate Center, University of Goettingen, Kellnerweg 4, 37077 Göttingen, Germany
| |
Collapse
|
28
|
Hellwege JN, Stallings SC, Piekos JA, Jasper EA, Aronoff DM, Edwards TL, Velez Edwards DR. Association of genetically-predicted placental gene expression with adult blood pressure traits. J Hypertens 2023; 41:1024-1032. [PMID: 37016918 PMCID: PMC10287061 DOI: 10.1097/hjh.0000000000003427] [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] [Indexed: 04/06/2023]
Abstract
OBJECTIVE Blood pressure is a complex, polygenic trait, and the need to identify prehypertensive risks and new gene targets for blood pressure control therapies or prevention continues. We hypothesize a developmental origins model of blood pressure traits through the life course where the placenta is a conduit mediating genomic and nongenomic transmission of disease risk. Genetic control of placental gene expression has recently been described through expression quantitative trait loci (eQTL) studies which have identified associations with childhood phenotypes. METHODS We conducted a transcriptome-wide gene expression analysis estimating the predicted gene expression of placental tissue in adult individuals with genome-wide association study (GWAS) blood pressure summary statistics. We constructed predicted expression models of 15 154 genes from reference placenta eQTL data and investigated whether genetically-predicted gene expression in placental tissue is associated with blood pressure traits using published GWAS summary statistics. Functional annotation of significant genes was generated using FUMA. RESULTS We identified 18, 9, and 21 genes where predicted expression in placenta was significantly associated with systolic blood pressure (SBP), diastolic blood pressure (DBP), and pulse pressure (PP), respectively. There were 14 gene-tissue associations (13 unique genes) significant only in placenta. CONCLUSIONS In this meta-analysis using S-PrediXcan and GWAS summary statistics, the predicted expression in placenta of 48 genes was statistically significantly associated with blood pressure traits. Notable findings included the association of FGFR1 expression with increased SBP and PP. This evidence of gene expression variation in placenta preceding the onset of adult blood pressure phenotypes is an example of extreme preclinical biological changes which may benefit from intervention.
Collapse
Affiliation(s)
- Jacklyn N Hellwege
- Department of Medicine, Division of Genetic Medicine
- Vanderbilt Genetics Institute
| | - Sarah C Stallings
- Department of Medicine, Division of Genetic Medicine
- Vanderbilt Genetics Institute
| | - Jacqueline A Piekos
- Vanderbilt Genetics Institute
- Department of Obstetrics and Gynecology, Division of Quantitative Sciences
| | - Elizabeth A Jasper
- Department of Obstetrics and Gynecology, Division of Quantitative Sciences
| | - David M Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Todd L Edwards
- Vanderbilt Genetics Institute
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute
- Department of Obstetrics and Gynecology, Division of Quantitative Sciences
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Richard D, Capellini TD, Diekman BO. Epigenetics as a mediator of genetic risk in osteoarthritis: role during development, homeostasis, aging, and disease progression. Am J Physiol Cell Physiol 2023; 324:C1078-C1088. [PMID: 36971423 PMCID: PMC10191130 DOI: 10.1152/ajpcell.00574.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
The identification of genomic loci that are associated with osteoarthritis (OA) has provided a starting point for understanding how genetic variation activates catabolic processes in the joint. However, genetic variants can only alter gene expression and cellular function when the epigenetic environment is permissive to these effects. In this review, we provide examples of how epigenetic shifts at distinct life stages can alter the risk for OA, which we posit is critical for the proper interpretation of genome-wide association studies (GWAS). During development, intensive work on the growth and differentiation factor 5 (GDF5) locus has revealed the importance of tissue-specific enhancer activity in controlling both joint development and the subsequent risk for OA. During homeostasis in adults, underlying genetic risk factors may help establish beneficial or catabolic "set points" that dictate tissue function, with a strong cumulative effect on OA risk. During aging, methylation changes and the reorganization of chromatin can "unmask" the effects of genetic variants. The destructive function of variants that alter aging would only mediate effects after reproductive competence and thus avoid any evolutionary selection pressure, as consistent with larger frameworks of biological aging and its relationship to disease. A similar "unmasking" may occur during OA progression, which is supported by the finding of distinct expression quantitative trait loci (eQTLs) in chondrocytes depending on the degree of tissue degradation. Finally, we propose that massively parallel reporter assays (MPRAs) will be a valuable tool to test the function of putative OA GWAS variants in chondrocytes from different life stages.
Collapse
Affiliation(s)
- Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, United States
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
30
|
Fortier I, Wey TW, Bergeron J, Pinot de Moira A, Nybo-Andersen AM, Bishop T, Murtagh MJ, Miočević M, Swertz MA, van Enckevort E, Marcon Y, Mayrhofer MT, Ornelas JP, Sebert S, Santos AC, Rocha A, Wilson RC, Griffith LE, Burton P. Life course of retrospective harmonization initiatives: key elements to consider. J Dev Orig Health Dis 2023; 14:190-198. [PMID: 35957574 DOI: 10.1017/s2040174422000460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Optimizing research on the developmental origins of health and disease (DOHaD) involves implementing initiatives maximizing the use of the available cohort study data; achieving sufficient statistical power to support subgroup analysis; and using participant data presenting adequate follow-up and exposure heterogeneity. It also involves being able to undertake comparison, cross-validation, or replication across data sets. To answer these requirements, cohort study data need to be findable, accessible, interoperable, and reusable (FAIR), and more particularly, it often needs to be harmonized. Harmonization is required to achieve or improve comparability of the putatively equivalent measures collected by different studies on different individuals. Although the characteristics of the research initiatives generating and using harmonized data vary extensively, all are confronted by similar issues. Having to collate, understand, process, host, and co-analyze data from individual cohort studies is particularly challenging. The scientific success and timely management of projects can be facilitated by an ensemble of factors. The current document provides an overview of the 'life course' of research projects requiring harmonization of existing data and highlights key elements to be considered from the inception to the end of the project.
Collapse
Affiliation(s)
- Isabel Fortier
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Tina W Wey
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Julie Bergeron
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | | | | - Tom Bishop
- Epidemiology Unit, University of Cambridge, England, UK
| | - Madeleine J Murtagh
- School of Social and Political Sciences, University of Glasgow, Scotland, UK
| | - Milica Miočević
- Department of Psychology, McGill University, Montreal, QC, Canada
| | - Morris A Swertz
- University Medical Center Groningen, University of Groningen, Netherlands
| | - Esther van Enckevort
- Department of Genetics, University Medical Center Groningen, University of Groningen, Netherlands
| | | | | | - Jos Pedro Ornelas
- INESC TEC - Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
| | | | - Ana Cristina Santos
- Department of Epidemiology, Institute of Public Health of the University of Porto, Portugal
| | - Artur Rocha
- INESC TEC - Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
| | - Rebecca C Wilson
- Department of Public Health, Policy and Systems, University of Liverpool, Liverpool, England, UK
| | - Lauren E Griffith
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Paul Burton
- Population Health Sciences Institute, Newcastle University, Newcastle-upon-Tyne, England, UK
| |
Collapse
|
31
|
Meka K, Jacob CM, Modi N, Bustreo F, Di Renzo GC, Malamitsi-Puchner A, Briana D, Iaia DG, Fogstad H, Tamvada JP, Moreno IO, Hanson M. Valuing maternal, newborn, child and adolescent health for societal progress - Going beyond the economic orthodoxy of gross domestic product. Acta Paediatr 2023; 112:630-634. [PMID: 36656112 DOI: 10.1111/apa.16677] [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: 12/19/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
AIM In view of the long-standing recognition that gross domestic product (GDP) does not capture the unremunerated work largely conducted by women upon which societal well-being depends, to discuss the implications for GDP of maternal, newborn, child and adolescent health (MNCAH), and its influences on health, well-being and prosperity across the life course and across generations. METHODS A wide-ranging discussion of the informal think-tank The Venice Forum was held over two days, with inputs from invited experts in person and online. RESULTS There was consensus that a strong case could be made for inclusion of unremunerated work largely conducted by women as a positive contribution to GDP in view of its impact on future health and prosperity, and conversely exclusion from GDP of outputs from industries which harm health. CONCLUSION Taken with the current challenges from COVID, climate change and conflict, there is a compelling need to redefine economic progress through equitable models and metrics that incorporate short-/medium-/long-term societal value of activities that improve MNCAH.
Collapse
Affiliation(s)
- Kiran Meka
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Chandni M Jacob
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Southampton NIHR Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Neena Modi
- Section of Neonatal Medicine, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Flavia Bustreo
- Partnership for Maternal, Newborn and Child Health Board, Geneva, Switzerland
| | - Gian Carlo Di Renzo
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy.,Department of Obstetrics and Gynaecology, I.M. Sechenov First State University of Moscow, Moscow, Russia
| | - Ariadne Malamitsi-Puchner
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Briana
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Domenico G Iaia
- Partnership for Maternal Newborn and Child Health, World Health Organisation, Geneva, Switzerland
| | - Helga Fogstad
- Partnership for Maternal Newborn and Child Health, World Health Organisation, Geneva, Switzerland
| | - Jagannadha P Tamvada
- Southampton Business School, Faculty of Social Sciences, University of Southampton, Southampton, UK
| | - Ivan Ochoa Moreno
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mark Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Southampton NIHR Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
32
|
Kurihara C, Kuniyoshi KM, Rehan VK. Preterm Birth, Developmental Smoke/Nicotine Exposure, and Life-Long Pulmonary Sequelae. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040608. [PMID: 37189857 DOI: 10.3390/children10040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
This review delineates the main pulmonary issues related to preterm birth, perinatal tobacco/nicotine exposure, and its effects on offspring, focusing on respiratory health and its possible transmission to subsequent generations. We review the extent of the problem of preterm birth, prematurity-related pulmonary effects, and the associated increased risk of asthma later in life. We then review the impact of developmental tobacco/nicotine exposure on offspring asthma and the significance of transgenerational pulmonary effects following perinatal tobacco/nicotine exposure, possibly via its effects on germline epigenetics.
Collapse
Affiliation(s)
- Chie Kurihara
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Katherine M Kuniyoshi
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Virender K Rehan
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Saedi S, Watson SE, Young JL, Tan Y, Wintergerst KA, Cai L. Does maternal low-dose cadmium exposure increase the risk of offspring to develop metabolic syndrome and/or type 2 diabetes? Life Sci 2023; 315:121385. [PMID: 36634865 PMCID: PMC9912173 DOI: 10.1016/j.lfs.2023.121385] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/31/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Cadmium is a hazardous metal with multiple organ toxicity that causes great harm to human health. Cadmium enters the human body through occupational exposure, diet, drinking water, breathing, and smoking. Cadmium accumulation in the human body is associated with increased risk of developing obesity, cardiovascular disease, diabetes, and metabolic syndrome (MetS). Cadmium uptake is enhanced during pregnancy and can cross the placenta affecting placental development and function. Subsequently, cadmium can pass to fetus, gathering in multiple organs such as the liver and pancreas. Early-life cadmium exposure can induce hepatic oxidative stress and pancreatic β-cell dysfunction, resulting in insulin resistance and glucose metabolic dyshomeostasis in the offspring. Prenatal exposure to cadmium is also associated with increasing epigenetic effects on the offspring's multi-organ functions. However, whether and how maternal exposure to low-dose cadmium impacts the risks of developing type 2 diabetes (T2D) in the young and/or adult offspring remains unclear. This review collected available data to address the current evidence for the potential role of cadmium exposure, leading to insulin resistance and the development of T2D in offspring. However, this review reveals that underlying mechanisms linking prenatal cadmium exposure during pregnancy with T2D in offspring remain to be adequately investigated.
Collapse
Affiliation(s)
- Saman Saedi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Sara E Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
| | - Jamie L Young
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Hospital, Louisville, KY, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA; Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
34
|
Pappalardo XG, Testa G, Pellitteri R, Dell’Albani P, Rodolico M, Pavone V, Parano E. Early Life Stress (ELS) Effects on Fetal and Adult Bone Development. CHILDREN (BASEL, SWITZERLAND) 2023; 10:102. [PMID: 36670652 PMCID: PMC9856960 DOI: 10.3390/children10010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023]
Abstract
Early life stress (ELS) refers to harmful environmental events (i.e., poor maternal health, metabolic restraint, childhood trauma) occurring during the prenatal and/or postnatal period, which may cause the 'epigenetic corruption' of cellular and molecular signaling of mental and physical development. While the impact of ELS in a wide range of human diseases has been confirmed, the ELS susceptibility to bone diseases has been poorly explored. In this review, to understand the potential mediating pathways of ELS in bone diseases, PRISMA criteria were used to analyze different stress protocols in mammal models and the effects elicited in dams and their progeny. Data collected, despite the methodological heterogeneity, show that ELS interferes with fetal bone formation, also revealing that the stress type and affected developmental phase may influence the variety and severity of bone anomalies. Interestingly, these findings highlight the maternal and fetal ability to buffer stress, establishing a new role for the placenta in minimizing ELS perturbations. The functional link between ELS and bone impairments will boost future investigations on maternal stress transmission to the fetus and, parallelly, help the assessment of catch-up mechanisms of skeleton adaptations from the cascading ELS effects.
Collapse
Affiliation(s)
- Xena Giada Pappalardo
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Gianluca Testa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, Hospital Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy
| | - Rosalia Pellitteri
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Paola Dell’Albani
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Margherita Rodolico
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Vito Pavone
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, Hospital Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy
| | - Enrico Parano
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| |
Collapse
|
35
|
Castaño-Moreno E, Ronco AM, Casanello P. Metabolic Interaction Between Folate, Vitamin B12, and Polyunsaturated Fatty Acids in Pregnancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:83-98. [PMID: 37466770 DOI: 10.1007/978-3-031-32554-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Fetal growth and development are influenced by maternal nutrition and gestational weight gain. Adequate intake of nutrients such as folate, vitamin B12, and docosahexaenoic acid (DHA) is essential for healthy fetal and placental development. Many countries have a national flour fortification program with folic acid (FA), together with pre-pregnancy supplementation of FA (400 μg/day) during the first trimester of pregnancy. The latter has been recommended by the WHO and adapted to local requirements by perinatal guidelines. On the other hand, in population studies, many women of childbearing age have vitamin B12 deficiency (<148 pmol/L), which can be additionally masked by high FA intake and maternal pregestational obesity. Under these conditions, these patients could be having pregnancies in a folate/vitamin B12 imbalance, which is associated with higher adiposity, insulin resistance, altered lipid metabolism, and low DHA levels in their offspring. However, if these neonatal consequences of maternal pregestational obesity and folate/vitamin B12 imbalance can be reverted by DHA supplementation during pregnancy has not been addressed. This chapter reviews the literature and exposes the current gaps in knowledge and challenges in maternal nutrition with a life-course perspective.
Collapse
Affiliation(s)
- Erika Castaño-Moreno
- Laboratorio de Nutrición y Regulación Metabólica, Unidad de Nutrición Humana - Instituto de Nutrición y Tecnología de los Alimentos (INTA) - Universidad de Chile, Santiago, Chile
- Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, Mexico
| | - Ana María Ronco
- Laboratorio de Nutrición y Regulación Metabólica, Unidad de Nutrición Humana - Instituto de Nutrición y Tecnología de los Alimentos (INTA) - Universidad de Chile, Santiago, Chile
| | - Paola Casanello
- Department of Neonatology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
36
|
Abruzzese GA, Arbocco FCV, Ferrer MJ, Silva AF, Motta AB. Role of Hormones During Gestation and Early Development: Pathways Involved in Developmental Programming. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:31-70. [PMID: 37466768 DOI: 10.1007/978-3-031-32554-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Accumulating evidence suggests that an altered maternal milieu and environmental insults during the intrauterine and perinatal periods of life affect the developing organism, leading to detrimental long-term outcomes and often to adult pathologies through programming effects. Hormones, together with growth factors, play critical roles in the regulation of maternal-fetal and maternal-neonate interfaces, and alterations in any of them may lead to programming effects on the developing organism. In this chapter, we will review the role of sex steroids, thyroid hormones, and insulin-like growth factors, as crucial factors involved in physiological processes during pregnancy and lactation, and their role in developmental programming effects during fetal and early neonatal life. Also, we will consider epidemiological evidence and data from animal models of altered maternal hormonal environments and focus on the role of different tissues in the establishment of maternal and fetus/infant interaction. Finally, we will identify unresolved questions and discuss potential future research directions.
Collapse
Affiliation(s)
- Giselle Adriana Abruzzese
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Fiorella Campo Verde Arbocco
- Laboratorio de Hormonas y Biología del Cáncer, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, Mendoza, Argentina
- Laboratorio de Reproducción y Lactancia, IMBECU, CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| | - María José Ferrer
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Aimé Florencia Silva
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Alicia Beatriz Motta
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
37
|
Bishop AC, Spradling‐Reeves KD, Shade RE, Lange KJ, Birnbaum S, Favela K, Dick EJ, Nijland MJ, Li C, Nathanielsz PW, Cox LA. Postnatal persistence of nonhuman primate sex-dependent renal structural and molecular changes programmed by intrauterine growth restriction. J Med Primatol 2022; 51:329-344. [PMID: 35855511 PMCID: PMC9796938 DOI: 10.1111/jmp.12601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Poor nutrition during fetal development programs postnatal kidney function. Understanding postnatal consequences in nonhuman primates (NHP) is important for translation to our understanding the impact on human kidney function and disease risk. We hypothesized that intrauterine growth restriction (IUGR) in NHP persists postnatally, with potential molecular mechanisms revealed by Western-type diet challenge. METHODS IUGR juvenile baboons were fed a 7-week Western diet, with kidney biopsies, blood, and urine collected before and after challenge. Transcriptomics and metabolomics were used to analyze biosamples. RESULTS Pre-challenge IUGR kidney transcriptome and urine metabolome differed from controls. Post-challenge, sex and diet-specific responses in urine metabolite and renal signaling pathways were observed. Dysregulated mTOR signaling persisted postnatally in female pre-challenge. Post-challenge IUGR male response showed uncoordinated signaling suggesting proximal tubule injury. CONCLUSION Fetal undernutrition impacts juvenile offspring kidneys at the molecular level suggesting early-onset blood pressure dysregulation.
Collapse
Affiliation(s)
- Andrew C. Bishop
- Center for Precision MedicineDepartment of Internal Medicine, Wake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kimberly D. Spradling‐Reeves
- Center for Precision MedicineDepartment of Internal Medicine, Wake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Robert E. Shade
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Kenneth J. Lange
- Department of Pharmaceuticals and BioengineeringSouthwest Research InstituteSan AntonioTexasUSA
| | - Shifra Birnbaum
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Kristin Favela
- Department of Pharmaceuticals and BioengineeringSouthwest Research InstituteSan AntonioTexasUSA
| | - Edward J. Dick
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Mark J. Nijland
- Department of Obstetrics and GynecologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Cun Li
- Department of Animal SciencesUniversity of WyomingLaramieWyomingUSA
| | - Peter W. Nathanielsz
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTexasUSA
- Department of Animal SciencesUniversity of WyomingLaramieWyomingUSA
| | - Laura A. Cox
- Center for Precision MedicineDepartment of Internal Medicine, Wake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTexasUSA
| |
Collapse
|
38
|
Adaptation of Helicoverpa armigera to Soybean Peptidase Inhibitors Is Associated with the Transgenerational Upregulation of Serine Peptidases. Int J Mol Sci 2022; 23:ijms232214301. [PMID: 36430785 PMCID: PMC9693090 DOI: 10.3390/ijms232214301] [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: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
Molecular phenotypes induced by environmental stimuli can be transmitted to offspring through epigenetic inheritance. Using transcriptome profiling, we show that the adaptation of Helicoverpa armigera larvae to soybean peptidase inhibitors (SPIs) is associated with large-scale gene expression changes including the upregulation of genes encoding serine peptidases in the digestive system. Furthermore, approximately 60% of the gene expression changes induced by SPIs persisted in the next generation of larvae fed on SPI-free diets including genes encoding regulatory, oxidoreductase, and protease functions. To investigate the role of epigenetic mechanisms in regulating SPI adaptation, the methylome of the digestive system of first-generation larvae (fed on a diet with and without SPIs) and of the progeny of larvae exposed to SPIs were characterized. A comparative analysis between RNA-seq and Methyl-seq data did not show a direct relationship between differentially methylated and differentially expressed genes, while trypsin and chymotrypsin genes were unmethylated in all treatments. Rather, DNA methylation potential epialleles were associated with transcriptional and translational controls; these may play a regulatory role in the adaptation of H. armigera to SPIs. Altogether, our findings provided insight into the mechanisms of insect adaptation to plant antiherbivore defense proteins and illustrated how large-scale transcriptional reprograming of insect genes can be transmitted across generations.
Collapse
|
39
|
Using a Very Low Energy Diet to Achieve Substantial Preconception Weight Loss in Women with Obesity: A Review of the Safety and Efficacy. Nutrients 2022; 14:nu14204423. [PMID: 36297107 PMCID: PMC9608905 DOI: 10.3390/nu14204423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/29/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity in women of reproductive age is common. Emerging evidence suggests that maternal obesity not only increases the risk of adverse pregnancy outcomes but also has an enduring impact on the metabolic health of the offspring. Given this, management of obesity prior to pregnancy is critically important. Almost all international guidelines suggest that women with obesity should aim to achieve weight loss prior to pregnancy. However, current pre-conception weight loss therapies are sub-optimal. Lifestyle modification typically results in modest weight loss. This may assist fertility but does not alter pregnancy outcomes. Bariatric surgery results in substantial weight loss, which improves pregnancy outcomes for the mother but may be harmful to the offspring. Alternative approaches to the management of obesity in women planning pregnancy are needed. Very low energy diets (VLEDs) have been proposed as a possible tool to assist women with obesity achieve weight loss prior to conception. While VLEDs can induce substantial and rapid weight loss, there are concerns about the impact of rapid weight loss on maternal nutrition prior to pregnancy and about inadvertent exposure of the early fetus to ketosis. The purpose of this review is to examine the existing literature regarding the safety and efficacy of a preconception VLED program as a tool to achieve substantial weight loss in women with obesity.
Collapse
|
40
|
Lee SH, Liu X, Jimenez-Morales D, Rinaudo PF. Murine blastocysts generated by in vitro fertilization show increased Warburg metabolism and altered lactate production. eLife 2022; 11:e79153. [PMID: 36107481 PMCID: PMC9519152 DOI: 10.7554/elife.79153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
In vitro fertilization (IVF) has resulted in the birth of over 8 million children. Although most IVF-conceived children are healthy, several studies suggest an increased risk of altered growth rate, cardiovascular dysfunction, and glucose intolerance in this population compared to naturally conceived children. However, a clear understanding of how embryonic metabolism is affected by culture condition and how embryos reprogram their metabolism is unknown. Here, we studied oxidative stress and metabolic alteration in blastocysts conceived by natural mating or by IVF and cultured in physiologic (5%) or atmospheric (20%) oxygen. We found that IVF-generated blastocysts manifest increased reactive oxygen species, oxidative damage to DNA/lipid/proteins, and reduction in glutathione. Metabolic analysis revealed IVF-generated blastocysts display decreased mitochondria respiration and increased glycolytic activity suggestive of enhanced Warburg metabolism. These findings were corroborated by altered intracellular and extracellular pH and increased intracellular lactate levels in IVF-generated embryos. Comprehensive proteomic analysis and targeted immunofluorescence showed reduction of lactate dehydrogenase-B and monocarboxylate transporter 1, enzymes involved in lactate metabolism. Importantly, these enzymes remained downregulated in the tissues of adult IVF-conceived mice, suggesting that metabolic alterations in IVF-generated embryos may result in alteration in lactate metabolism. These findings suggest that alterations in lactate metabolism are a likely mechanism involved in genomic reprogramming and could be involved in the developmental origin of health and disease.
Collapse
Affiliation(s)
- Seok Hee Lee
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California, San FranciscoSan FranciscoUnited States
| | - Xiaowei Liu
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California, San FranciscoSan FranciscoUnited States
| | - David Jimenez-Morales
- Division of Cardiovascular Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Paolo F Rinaudo
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
41
|
Jones MA, Whitaker K, Taverno Ross SE, Davis K, Libertus K, Gibbs BB. Maternal Sedentary Behavior and Physical Activity across Pregnancy and Early Childhood Growth. Child Obes 2022; 18:399-408. [PMID: 35108109 PMCID: PMC9634982 DOI: 10.1089/chi.2021.0202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Evidence suggests in utero exposures are related to lifespan health of the offspring. Whether maternal activity profile during pregnancy impacts offspring health remains unknown. Methods: This follow-up study recruited mothers with objectively measured sedentary behavior (SED) and moderate-to-vigorous intensity physical activity (MVPA) from a previous cohort study. Maternal activity was analyzed across pregnancy (trajectory groups) and continuously by trimester. Offspring anthropometrics up to 24 months were abstracted from medical records (n = 62). Outcomes included childhood growth rate (incremental rate of BMI z-score change up to 24 months) and rapid growth (increased BMI z-score >0.67 at 12 months). Associations of maternal activity with growth rate were examined using mixed linear models and rapid growth using generalized linear models. Results: Forty percent of participants were in the high SED and 20% in the high MVPA trajectories during pregnancy. Higher SED, across pregnancy [slope (95% confidence interval; CI): 0.080 (0.024-0.061) ΔBMI z-score/month] and in the first trimester [standardized beta; std β (95% CI): 0.017 (0.007-0.026)], was related to accelerated growth rate. Higher MVPA, in the second and third trimesters, was associated with accelerated growth rate [std β (95% CI): trimester 2: 0.013 (0.002-0.024) and trimester 3:0.011 (0.003-0.020)] and greater risk of rapid growth [risk ratio (95% CI): trimester 2: 1.25 (1.009-1.555) and trimester 3: 1.25 (1.056-1.475)]. Conclusions: These findings add to growing evidence on the deleterious effects of high SED during pregnancy. The increased risk for accelerated growth with higher MVPA elicits further investigation. Overall, maternal activity profile shows promise as a modifiable behavior to improve intergenerational health.
Collapse
Affiliation(s)
- Melissa A. Jones
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA
| | - Kara Whitaker
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA
| | - Sharon E. Taverno Ross
- Department of Health and Human Development and University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelliann Davis
- Department of Health and Human Development and University of Pittsburgh, Pittsburgh, PA, USA
| | - Klaus Libertus
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bethany Barone Gibbs
- Department of Health and Human Development and University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
42
|
Newborn screen metabolic panels reflect the impact of common disorders of pregnancy. Pediatr Res 2022; 92:490-497. [PMID: 34671094 PMCID: PMC10265936 DOI: 10.1038/s41390-021-01753-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Hypertensive disorders of pregnancy and maternal diabetes profoundly affect fetal and newborn growth, yet disturbances in intermediate metabolism and relevant mediators of fetal growth alterations remain poorly defined. We sought to determine whether there are distinct newborn screen metabolic patterns among newborns affected by maternal hypertensive disorders or diabetes in utero. METHODS A retrospective observational study investigating distinct newborn screen metabolites in conjunction with data linked to birth and hospitalization records in the state of California between 2005 and 2010. RESULTS A total of 41,333 maternal-infant dyads were included. Infants of diabetic mothers demonstrated associations with short-chain acylcarnitines and free carnitine. Infants born to mothers with preeclampsia with severe features and chronic hypertension with superimposed preeclampsia had alterations in acetylcarnitine, free carnitine, and ornithine levels. These results were further accentuated by size for gestational age designations. CONCLUSIONS Infants of diabetic mothers demonstrate metabolic signs of incomplete beta oxidation and altered lipid metabolism. Infants of mothers with hypertensive disorders of pregnancy carry analyte signals that may reflect oxidative stress via altered nitric oxide signaling. The newborn screen analyte composition is influenced by the presence of these maternal conditions and is further associated with the newborn size designation at birth. IMPACT Substantial differences in newborn screen analyte profiles were present based on the presence or absence of maternal diabetes or hypertensive disorder of pregnancy and this finding was further influenced by the newborn size designation at birth. The metabolic health of the newborn can be examined using the newborn screen and is heavily impacted by the condition of the mother during pregnancy. Utilizing the newborn screen to identify newborns affected by common conditions of pregnancy may help relate an infant's underlying biological disposition with their clinical phenotype allowing for greater risk stratification and intervention.
Collapse
|
43
|
Velasquez-Vasconez PA. Reflections about the Molecular Tool That Could Change the Course of Human History: Genome Editing. PERSONA Y BIOÉTICA 2022. [DOI: 10.5294/pebi.2022.26.1.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Genetic editing has many applications in almost all areas of society, but may also lead to unpredictable consequences. Genome editing to modify the human germline is at the center of global discussion. Owing to the increasing number of unanswered scientific, ethical, and policy questions, the scientific community agrees that it would be inappropriate to genetically modify embryos. A serious and open debate is necessary to decide whether such research should be suspended or encouraged. Here we show some bold arguments in favor of deleting deleterious genes from the human genome and the risks liberal eugenism poses.
Collapse
|
44
|
Faienza MF, Urbano F, Lassandro G, Valente F, D’Amato G, Portincasa P, Giordano P. The Cardiovascular Disease (CVD) Risk Continuum from Prenatal Life to Adulthood: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8282. [PMID: 35886134 PMCID: PMC9317926 DOI: 10.3390/ijerph19148282] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
The risk of developing cardiovascular diseases (CVDs) arises from the interaction of prenatal factors; epigenetic regulation; neonatal factors; and factors that affect childhood and adolescence, such as early adiposity rebound (AR) and social and environmental influences. Thus, CVD risk varies between the group of low-risk metabolically healthy normal-weight subjects (MHNW); the intermediate-risk group, which includes metabolically healthy obese (MHO) and metabolically unhealthy normal-weight subjects (MUHNW); and the high-risk group of metabolically unhealthy obese (MUHO) subjects. In this continuum, several risk factors come into play and contribute to endothelial damage, vascular and myocardial remodeling, and atherosclerotic processes. These pathologies can occur both in prenatal life and in early childhood and contribute to significantly increasing CVD risk in young adults over time. Early intervention in the pediatric MUHO population to reduce the CVD risk during adulthood remains a challenge. In this review, we focus on CVD risk factors arising at different stages of life by performing a search of the recent literature. It is urgent to focus on preventive or early therapeutic strategies to stop this disturbing negative metabolic trend, which manifests as a continuum from prenatal life to adulthood.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Flavia Urbano
- Giovanni XXIII Pediatric Hospital, 70126 Bari, Italy; (F.U.); (G.L.)
| | | | - Federica Valente
- Department of Cardiology, Erasme University Hospital, Université Libre de Bruxelles, 1050 Brussels, Belgium;
| | - Gabriele D’Amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70131 Bari, Italy;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari “A. Moro”, 70121 Bari, Italy;
| | - Paola Giordano
- Department of Interdisciplinary Medicine, Pediatric Unit, University of Bari “A. Moro”, 70121 Bari, Italy;
| |
Collapse
|
45
|
Ochoa‐Moreno I, Hanson M. Obesity and diabetes in pregnancy: a perfect storm for transgenerational health. PRACTICAL DIABETES 2022. [DOI: 10.1002/pdi.2403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Ivan Ochoa‐Moreno
- Institute of Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton Southampton UK
| | - Mark Hanson
- Institute of Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton Southampton UK
- Southampton NIHR Biomedical Research Centre University Hospital Southampton UK
| |
Collapse
|
46
|
Olstad EW, Nordeng HME, Sandve GK, Lyle R, Gervin K. Low reliability of DNA methylation across Illumina Infinium platforms in cord blood: implications for replication studies and meta-analyses of prenatal exposures. Clin Epigenetics 2022; 14:80. [PMID: 35765087 PMCID: PMC9238140 DOI: 10.1186/s13148-022-01299-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/12/2022] [Indexed: 11/20/2022] Open
Abstract
Background There is an increasing interest in the role of epigenetics in epidemiology, but the emerging research field faces several critical biological and technical challenges. In particular, recent studies have shown poor correlation of measured DNA methylation (DNAm) levels within and across Illumina Infinium platforms in various tissues. In this study, we have investigated concordance between 450 k and EPIC Infinium platforms in cord blood. We could not replicate our previous findings on the association of prenatal paracetamol exposure with cord blood DNAm, which prompted an investigation of cross-platform DNAm differences. Results This study is based on two DNAm data sets from cord blood samples selected from the Norwegian Mother, Father and Child Cohort Study (MoBa). DNAm of one data set was measured using the 450 k platform and the other data set was measured using the EPIC platform. Initial analyses of the EPIC data could not replicate any of our previous significant findings in the 450 k data on associations between prenatal paracetamol exposure and cord blood DNAm. A subset of the samples (n = 17) was included in both data sets, which enabled analyses of technical sources potentially contributing to the negative replication. Analyses of these 17 samples with repeated measurements revealed high per-sample correlations (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\stackrel{\mathrm{-}}{\text{R}}\hspace{0.17em}\approx$$\end{document}R-≈ 0.99), but low per-CpG correlations (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\stackrel{\mathrm{-}}{\text{R}}$$\end{document}R- ≈ 0.24) between the platforms. 1.7% of the CpGs exhibited a mean DNAm difference across platforms > 0.1. Furthermore, only 26.7% of the CpGs exhibited a moderate or better cross-platform reliability (intra-class correlation coefficient ≥ 0.5). Conclusion The observations of low cross-platform probe correlation and reliability corroborate previous reports in other tissues. Our study cannot determine the origin of the differences between platforms. Nevertheless, it emulates the setting in studies using data from multiple Infinium platforms, often analysed several years apart. Therefore, the findings may have important implications for future epigenome-wide association studies (EWASs), in replication, meta-analyses and longitudinal studies. Cognisance and transparency of the challenges related to cross-platform studies may enhance the interpretation, replicability and validity of EWAS results both in cord blood and other tissues, ultimately improving the clinical relevance of epigenetic epidemiology. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01299-3.
Collapse
|
47
|
Tsai MS, Newman C, Macdonald DW, Buesching CD. Adverse weather during in utero development is linked to higher rates of later-life herpesvirus reactivation in adult European badgers, Meles meles. ROYAL SOCIETY OPEN SCIENCE 2022; 9:211749. [PMID: 35582658 PMCID: PMC9091846 DOI: 10.1098/rsos.211749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/12/2022] [Indexed: 05/03/2023]
Abstract
Maternal immune and/or metabolic conditions relating to stress or nutritional status can affect in utero development among offspring with subsequent implications for later-life responses to infections. We used free-ranging European badgers as a host-pathogen model to investigate how prenatal weather conditions affect later-life herpesvirus genital tract reactivation. We applied a sliding window analysis of weather conditions to 164 samples collected in 2018 from 95 individuals born between 2005-2016. We test if the monthly mean and variation in rainfall and temperature experienced by their mother during the 12 months of delayed implantation and gestation prior to parturition subsequently affected individual herpes reactivation rates among these offspring. We identified four influential prenatal seasonal weather windows that corresponded with previously identified critical climatic conditions affecting badger survival, fecundity and body condition. These all occurred during the pre-implantation rather than the post-implantation period. We conclude that environmental cues during the in utero period of delayed implantation may result in changes that affect an individual's developmental programming against infection or viral reactivation later in life. This illustrates how prenatal adversity caused by environmental factors, such as climate change, can impact wildlife health and population dynamics-an interaction largely overlooked in wildlife management and conservation programmes.
Collapse
Affiliation(s)
- Ming-shan Tsai
- Department of Zoology, Wildlife Conservation Research Unit, University of Oxford, Recanati-Kaplan Centre, Abingdon Road, Tubney House, Tubney, Oxfordshire OX13 5QL, UK
| | - Chris Newman
- Department of Zoology, Wildlife Conservation Research Unit, University of Oxford, Recanati-Kaplan Centre, Abingdon Road, Tubney House, Tubney, Oxfordshire OX13 5QL, UK
- Cook's Lake Farming Forestry and Wildlife Inc (Ecological Consultancy), Queens County, Nova Scotia, Canada
| | - David W. Macdonald
- Department of Zoology, Wildlife Conservation Research Unit, University of Oxford, Recanati-Kaplan Centre, Abingdon Road, Tubney House, Tubney, Oxfordshire OX13 5QL, UK
| | - Christina D. Buesching
- Cook's Lake Farming Forestry and Wildlife Inc (Ecological Consultancy), Queens County, Nova Scotia, Canada
- Department of Biology, Irving K. Barber Faculty of Science, The University of British Columbia, Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
48
|
Rostami Dovom M, Noroozzadeh M, Mosaffa N, Piryaei A, Zadeh-Vakili A, Aabdollahifar MA, Rahmati M, Farhadi-Azar M, Ramezani Tehrani F. Maternal Exposure to D-galactose Reduces Ovarian Reserve in Female Rat Offspring Later in Life. Int J Endocrinol Metab 2022; 20:e123206. [PMID: 35993036 PMCID: PMC9383541 DOI: 10.5812/ijem-123206] [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: 02/06/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Embryonic life is critical for the formation of ovaries in mammals, and the intrauterine environment may affect ovarian reserve. OBJECTIVES The present study aimed to investigate the impact of prenatal D-galactose exposure on ovarian reserve in female rat offspring in their later lives. METHODS Ten pregnant Wistar rats were randomly divided into two groups. In one group, rats were fed with 35% D-galactose-enriched food from the third day to the end of pregnancy, and in the other group, rats were fed with a standard diet throughout pregnancy. Female offspring (prenatally galactose-exposed rats and non-exposed control rats) were examined in terms of hormonal levels [anti-Mullerian hormones (AMH), follicle-stimulating hormone (FSH), and estradiol (E2)] and ovarian histology at 45 - 50, 105 - 110, and 180 - 185 days of their age. RESULTS The number of primordial follicles significantly decreased time-dependently in prenatally galactose-exposed rats compared to controls (P-value = 0.002). In addition, decreases in AMH (3.25 vs. 7.5 ng/mL; P = 0.000) and E2 (7.9 vs. 19.5 pg/mL; P = 0.000) and increases in FSH (6.5 vs. 0.8 mIU/mL; P < 0.007) were observed in galactose-exposed rats compared to controls at 45 - 50 days of age. CONCLUSIONS Prenatal exposure to D-galactose negatively affects ovarian reserve in female rats in their later lives. However, further investigation is needed to confirm our findings and explore underlying mechanisms.
Collapse
Affiliation(s)
- Marzieh Rostami Dovom
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Azita Zadeh-Vakili
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Aabdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rahmati
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahbanoo Farhadi-Azar
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
van Weelden W, Seed PT, Antoun E, Godfrey KM, Kitaba NT, Lillycrop KA, Dalrymple KV, Sobczyńska-Malefora A, Painter RC, Poston L, White SL, Flynn AC. Folate and vitamin B12 status: associations with maternal glucose and neonatal DNA methylation sites related to dysglycaemia, in pregnant women with obesity. J Dev Orig Health Dis 2022; 13:168-176. [PMID: 33972006 PMCID: PMC7612506 DOI: 10.1017/s2040174421000246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Recent studies implicate maternal gestational diabetes mellitus (GDM) in differential methylation of infant DNA. Folate and vitamin B12 play a role in DNA methylation, and these vitamins may also influence GDM risk. The aims of this study were to determine folate and vitamin B12 status in obese pregnant women and investigate associations between folate and vitamin B12 status, maternal dysglycaemia and neonatal DNA methylation at cytosine-phosphate-guanine sites previously observed to be associated with dysglycaemia. Obese pregnant women who participated in the UK Pregnancies Better Eating and Activity Trial were included. Serum folate and vitamin B12 were measured at the oral glucose tolerance test (OGTT) visit. Cord blood DNA methylation was assessed using the Infinium MethylationEPIC BeadChip. Regression models with adjustment for confounders were used to examine associations. Of the 951 women included, 356 (37.4%) were vitamin B12 deficient, and 44 (4.6%) were folate deficient. Two-hundred and seventy-one women (28%) developed GDM. Folate and vitamin B12 concentrations were not associated with neonatal DNA methylation. Higher folate was positively associated with 1-h plasma glucose after OGTT (β = 0.031, 95% CI 0.001-0.061, p = 0.045). There was no relationship between vitamin B12 and glucose concentrations post OGTT or between folate or vitamin B12 and GDM. In summary, we found no evidence to link folate and vitamin B12 status with the differential methylation of neonatal DNA previously observed in association with dysglycaemia. We add to the evidence that folate status may be related to maternal glucose homoeostasis although replication in other maternal cohorts is required for validation.
Collapse
Affiliation(s)
- Wenneke van Weelden
- Amsterdam University Medical Center – location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul T. Seed
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London
| | - Elie Antoun
- Human Development and Health, Faculty of Medicine, University of Southampton, UK
| | - Keith M. Godfrey
- Human Development and Health, Faculty of Medicine, University of Southampton, UK
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Negusse T. Kitaba
- Human Development and Health, Faculty of Medicine, University of Southampton, UK
| | - Karen A. Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Kathryn V. Dalrymple
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London
| | - Agata Sobczyńska-Malefora
- Nutristasis Unit, Viapath, St. Thomas’ Hospital, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Rebecca C. Painter
- Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lucilla Poston
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London
| | - Sara L. White
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London
| | - Angela C. Flynn
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London
| |
Collapse
|
50
|
Beltrán-Sánchez H, Palloni A, Huangfu Y, McEniry M. Population-level impact of adverse early life conditions on adult healthy life expectancy in low- and middle-income countries. POPULATION STUDIES 2022; 76:19-36. [PMID: 34110269 PMCID: PMC8660937 DOI: 10.1080/00324728.2021.1933149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Evidence from theories of Developmental Origins of Health and Disease (DOHaD) suggests that experiencing adverse early life conditions subsequently leads to detrimental adult health outcomes. The bulk of empirical DOHaD literature does not consider the nature and magnitude of the impact of adverse early life conditions at the population level. In particular, it ignores the distortion of age and cohort patterns of adult health and mortality and the increased load of chronic illness and disability that ensues. In this paper, we use a microsimulation model combined with empirical estimates of incidence and prevalence of obesity, type 2 diabetes, and associated disability in low- and middle-income countries to assess the magnitude of delayed effects on adult healthy life expectancy and on compression (or expansion) of morbidity at older ages. The main goal is to determine if, in what ways, and to what extent delayed effects due to early conditions can influence cohorts' chronic illness and disability profiles.
Collapse
Affiliation(s)
- Hiram Beltrán-Sánchez
- Department of Community Health Sciences, Fielding School of Public Health and California Center for Population Research, UCLA
| | - Alberto Palloni
- Center for Demography and Ecology, University of Wisconsin-Madison. Consejo Superior de Investigaciones (CSIC), Madrid-Spain
| | - Yiyue Huangfu
- Center for Demography and Ecology, University of Wisconsin-Madison
| | - Mary McEniry
- Center for Demography and Health of Aging, University of Wisconsin-Madison
| |
Collapse
|