1
|
Zhang C, Guo Y, Liu Y, Liu K, Hu W, Wang H. Sperm miR-142-3p Reprogramming Mediates Paternal Pre-Pregnancy Caffeine Exposure-Induced Non-Alcoholic Steatohepatitis in Male Offspring Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405592. [PMID: 39291441 DOI: 10.1002/advs.202405592] [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: 05/22/2024] [Revised: 07/28/2024] [Indexed: 09/19/2024]
Abstract
Numerous studies have suggested a strong association between paternal adverse environmental exposure and increased disease susceptibility in offspring. However, the impact of paternal pre-pregnant caffeine exposure (PPCE) on offspring health remains unexplored. This study elucidates the sperm reprogramming mechanism and potential intervention targets for PPCE-induced non-alcoholic steatohepatitis (NASH) in offspring. Here, male rats are administrated caffeine (15-60 mg kg-1/d) by gavage for 8 weeks and then mated with females to produce offspring. This study finds that NASH with transgenerational inheritance occurred in PPCE adult offspring. Mechanistically, a reduction of miR-142-3p is implicated in the occurrence of NASH, characterized by hepatic lipid metabolism dysfunction and chronic inflammation through an increase in ACSL4. Conversely, overexpression of miR-142-3p mitigated these manifestations. The origin of reduced miR-142-3p levels is traced to hypermethylation in the miR-142-3p promoter region of parental sperm, induced by elevated corticosterone levels rather than by caffeine per se. Similar outcomes are confirmed in offspring conceived via in vitro fertilization using miR-142-3pKO sperm. Overall, this study provides the first evidence of transgenerational inheritance of NASH in PPCE offspring and identifies miR-142-3p as a potential therapeutic target for NASH induced by paternal environmental adversities.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yu Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yi Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Kexin Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Wen Hu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| |
Collapse
|
2
|
Dimasi CG, Darby JRT, Cho SKS, Saini BS, Holman SL, Meakin AS, Wiese MD, Macgowan CK, Seed M, Morrison JL. Reduced in utero substrate supply decreases mitochondrial abundance and alters the expression of metabolic signalling molecules in the fetal sheep heart. J Physiol 2024; 602:5901-5922. [PMID: 37996982 DOI: 10.1113/jp285572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
Babies born with fetal growth restriction (FGR) are at higher risk of developing cardiometabolic diseases across the life course. The reduction in substrate supply to the developing fetus that causes FGR not only alters cardiac growth and structure but may have deleterious effects on metabolism and function. Using a sheep model of placental restriction to induce FGR, we investigated key cardiac metabolic and functional markers that may be altered in FGR. We also employed phase-contrast magnetic resonance imaging MRI to assess left ventricular cardiac output (LVCO) as a measure of cardiac function. We hypothesized that signalling molecules involved in cardiac fatty acid utilisation and contractility would be impaired by FGR and that this would have a negative impact on LVCO in the late gestation fetus. Key glucose (GLUT4 protein) and fatty acid (FATP, CD36 gene expression) substrate transporters were significantly reduced in the hearts of FGR fetuses. We also found reduced mitochondrial numbers as well as abundance of electron transport chain complexes (complexes II and IV). These data suggest that FGR diminishes metabolic and mitochondrial capacity in the fetal heart; however, alterations were not correlated with fetal LVCO. Overall, these data show that FGR alters fetal cardiac metabolism in late gestation. If sustained ex utero, this altered metabolic profile may contribute to poor cardiac outcomes in FGR-born individuals after birth. KEY POINTS: Around the time of birth, substrate utilisation in the fetal heart switches from carbohydrates to fatty acids. However, the effect of fetal growth restriction (FGR) on this switch, and thus the ability of the fetal heart to effectively metabolise fatty acids, is not fully understood. Using a sheep model of early onset FGR, we observed significant downregulation in mRNA expression of fatty acid receptors CD36 and FABP in the fetal heart. FGR fetuses also had significantly lower cardiac mitochondrial abundance than controls. There was a reduction in abundance of complexes II and IV within the electron transport chain of the FGR fetal heart, suggesting altered ATP production. This indicates reduced fatty acid metabolism and mitochondrial function in the heart of the FGR fetus, which may have detrimental long-term implications and contribute to increased risk of cardiovascular disease later in life.
Collapse
Affiliation(s)
- Catherine G Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Steven K S Cho
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Brahmdeep S Saini
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ashley S Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Michael D Wiese
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Christopher K Macgowan
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mike Seed
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Mobedi E, Dehghan Harati HR, Allahyari I, Gharagozlou F, Vojgani M, Baghbanani RH, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: V. Association of the main and interactive effects of maternal level of milk production and milk fat to protein ratio with offspring's birth weight, survival, and productive and reproductive performance from birth to the first lactation period. Theriogenology 2024; 228:17-29. [PMID: 39084064 DOI: 10.1016/j.theriogenology.2024.07.019] [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: 03/29/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Level of dam milk production (DMP) and dam milk fat to protein ratio (DFPR), as an indicator of metabolic status in dairy cows, have been identified to be associated with productive and reproductive performance of the offspring. Yet whether the interaction of DMP by DFPR can be associated with performance of the offspring have not been studied to our knowledge. Therefore, the present study was conducted to investigate the association of the main and interactive effects of DMP and DFPR with offspring's birth weight, survival, milk yield and fertility. To this end, data of birth weight, culling rate, milk yield and reproductive variables of offspring born to lactating dams (n = 14,582) and data associated with DMP and DFPR during 305-day lactation were retrieved. Afterwards, offspring were classified in three categories of DMP, including DMP1 (dams with <10.00 × 103 kg of 305-day milk production), DMP2 (dams with ≥10.00 × 103 kg and <14.00 × 103 kg of 305-day milk production), DMP3 (dams with ≥14.00 × 103 kg of 305-day milk production), and three categories of DFPR, including DFPR1 (offspring born to dams with <1.00 FPR), DFPR2 (offspring born to dams with ≥1.00 and < 1.40 FPR) and DFPR3 (offspring born to dams with ≥1.40 FPR). Statistical analysis revealed no association of the interaction effect of DMP by DFPR with investigated variables in the offspring (P > 0.05). However, the main effect of DMP was positively associated with milk yield, but negatively associated with survival, age at first insemination and conception during nulliparity, and transgenerational improvement in milk yield in the offspring (P < 0.05). Moreover, the main effect of DFPR was positively associated with birth weight, survival and first service conception rate during nulliparity, but negatively associated with metabolic status and reproductive performance during primiparity in the offspring (P < 0.05). In conclusion, the present study did not find any interaction effect of DMP by DFPR on productive and reproductive variables in the offspring. This finding implicates the association of DMP with milk production in the offspring was regardless of DFPR. Moreover, this finding implies the association of DFPR with postpartum metabolic status and reproductive performance in the offspring was regardless of DMP.
Collapse
Affiliation(s)
- Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
4
|
Yadav P, Mishra JS, Hurt MW, Chen DB, Kumar S. H2S donor GYY4137 mitigates sFlt-1-induced hypertension and vascular dysfunction in pregnant rats†. Biol Reprod 2024; 111:879-889. [PMID: 38938086 PMCID: PMC11473916 DOI: 10.1093/biolre/ioae103] [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: 03/18/2024] [Revised: 05/16/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024] Open
Abstract
Gestational hypertension, often associated with elevated soluble Fms-related receptor tyrosine kinase 1 (sFlt-1), poses significant risks to both maternal and fetal health. Hydrogen sulfide (H2S), a gasotransmitter, has demonstrated blood pressure-lowering effects in hypertensive animals and humans. However, its role in pregnancy-induced hypertension remains unclear. This study investigated the impact of GYY4137, a slow-release H2S donor, on sFlt-1-induced hypertension in pregnant rats . Pregnant rats were administered sFlt-1 (6 μg/kg/day, intravenously) or vehicle from gestation day (GD) 12-20. A subset of these groups received GYY4137 ( 50 mg/kg/day, intraperitoneal) from GD 16-20. Serum H2S levels, mean arterial blood pressure, uterine artery blood flow, and vascular reactivity were assessed. Elevated sFlt-1 reduced both maternal weight gain and serum H2S levels. GYY4137 treatment restored both weight gain and H2S levels in sFlt-1 dams. sFlt-1 increased mean arterial pressure and decreased uterine artery blood flow in pregnant rats. However, treatment with GYY4137 normalized blood pressure and restored uterine blood flow in sFlt-1 dams. sFlt-1 dams exhibited heightened vasoconstriction to phenylephrine and GYY4137 significantly mitigated the exaggerated vascular contraction. Notably, sFlt-1 impaired endothelium-dependent relaxation, while GYY4137 attenuated this impairment by upregulating eNOS protein levels and enhancing vasorelaxation in uterine arteries. GYY4137 mitigated sFlt-1-induced fetal growth restriction. In conclusion, sFlt-1 mediated hypertension is associated with decreased H2S levels. Replenishing H2S with the donor GYY4137 mitigates hypertension and improves vascular function and fetal growth outcomes. This suggests modulation of H2S could offer a novel therapeutic strategy for managing gestational hypertension and adverse fetal effects.
Collapse
Affiliation(s)
- Pankaj Yadav
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Mason William Hurt
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California, Irvine, CA, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
5
|
Huang M, Zeng X, Dai Z, Huang Y, Luo C, Tan X, Jiang Z, Fang X, Xu Y. Association between early exposure to famine and risk of renal impairment in adulthood: a systematic review and meta-analysis. Nutr Diabetes 2024; 14:84. [PMID: 39384564 PMCID: PMC11464504 DOI: 10.1038/s41387-024-00342-x] [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: 01/04/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/11/2024] Open
Abstract
Malnutrition early in life increases the later-life risk of noncommunicable diseases, and previous epidemiologic studies have found a link between famine and renal impairment, but no consensus has been reached. This meta-analysis and systematic review were conducted to assess the correlation between early-life famine exposure and the risk of developing renal impairment. Search in Embase, Scopus, Web of Science, PubMed, and Cochrane using keywords that report the correlation between early famine exposure and renal function indicators. RevMan and Stata software were used for data analysis. This meta-analysis contained twelve observational studies. The findings demonstrated a link between prenatal famine exposure and a higher risk of developing chronic kidney disease (CKD) (odds ratio (OR) = 1.73, 95% confidence interval (CI): 1.25, 2.39), a decreased estimated glomerular filtration rate (eGFR) (mean difference (MD) = -10.05, 95% CI: -11.64, -8.46), and increased serum creatinine (Scr) (MD = 0.02, 95% CI: 0.01, 0.03) compared to unexposed individuals. Famine exposure in childhood was associated with decreased eGFR (MD = -9.43, 95% CI: -12.01, -6.84) and increased Scr (MD = 0.03, 95% CI: 0.01, 0.04), but not with CKD (OR = 0.980, 95% CI: 0.53, 1.81). Famine exposure in adolescence and adulthood was associated with decreased eGFR (MD = -20.73, 95% CI: -22.40, -19.06). Evidence certainty was deemed to be of low or extremely low quality. Famine exposure early in life could pose a greater risk of developing renal impairment in adulthood, but this outcome may be driven by uncontrolled age differences between famine-births and post-famine-births (unexposed).
Collapse
Affiliation(s)
- Mengting Huang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xin Zeng
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Zhuojun Dai
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Yuqing Huang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Changfang Luo
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Zongzhe Jiang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xia Fang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China.
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Cilvik SN, Boehmer B, Wesolowski SR, Brown LD, Rozance PJ. Chronic late gestation fetal hyperglucagonaemia results in lower insulin secretion, pancreatic mass, islet area and beta- and α-cell proliferation. J Physiol 2024. [PMID: 39383208 DOI: 10.1113/jp286974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024] Open
Abstract
Fetal glucagon concentrations are elevated in the presence of a compromised intrauterine environment, as in cases of placental insufficiency and perinatal acidaemia. Our objective was to investigate the impact of late gestation fetal hyperglucagonaemia on in vivo insulin secretion and pancreatic islet structure. Chronically catheterized late gestation fetal sheep received an intravenous infusion of glucagon at low (5 ng/kg/min; GCG-5) or high (50 ng/kg/min; GCG-50) concentrations or a vehicle control (CON) for 8-10 days. Glucose-stimulated fetal insulin secretion (GSIS) was measured following 3 h (acute response) and 8-10 days (chronic response) of experimental infusions. Insulin, glucose and amino acid concentrations were measured longitudinally. The pancreas was collected at the study end for histology and gene expression analysis. Acute exposure (3 h) to GCG-50 induced a 3-fold increase in basal insulin concentrations with greater GSIS. Meanwhile, chronic exposure to both GCG-5 and GCG-50 decreased basal insulin concentrations 2-fold by day 8-10. Chronic GCG-50 also blunted GSIS at the study end. Fetal amino acid concentrations were decreased within 24 h of GCG-5 and GCG-50, while there were no differences in fetal glucose. Histologically, GCG-5 and GCG-50 had lower β- and α-cell proliferation, as well as lower α-cell mass and pancreas weight, while GCG-50 had lower islet area. This study demonstrates that chronic glucagon elevation in late gestation fetuses impairs β-cell proliferation and insulin secretion, which has the potential to contribute to later-life diabetes risk. We speculate that the action of glucagon in lower circulating fetal amino acid concentrations may have a suppressive effect on insulin secretion. KEY POINTS: We have previously demonstrated in a chronically catheterized fetal sheep model that experimentally elevated glucagon in the fetus impairs placental function, reduces fetal protein accretion and lowers fetal weight. In the present study, we further characterized the effects of elevated fetal glucagon on fetal physiology with a focus on pancreatic development and β-cell function. We show that experimentally elevated fetal glucagon results in lower β- and α-cell proliferation, as well as decreased insulin secretion after 8-10 days of glucagon infusion. These results have important implications for β-cell reserve and later-life predisposition to diabetes.
Collapse
Affiliation(s)
- Sarah N Cilvik
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brit Boehmer
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie R Wesolowski
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laura D Brown
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paul J Rozance
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
LeMoine FV, Hill H, Imbroane MR, Gautam AA, Van Dorn CH, Ranzini AC. Neonatal birthweight prediction using two- and three-dimensional estimated fetal weight among borderline small fetuses. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024. [PMID: 39368010 DOI: 10.1002/jcu.23844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/14/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE This study aimed (1) to determine the degree of correlation between 2D and 3D estimated fetal weight (EFW) and neonatal birth weight (BW) among borderline small fetuses and (2) to compare the accuracy and precision of 2D and 3D EFW in BW prediction. METHODS A retrospective cohort study evaluated fetuses who had an ultrasound performed between January 2017 and September 2021 at a tertiary maternal center. All singleton pregnancies with 3D EFW within 4 weeks of delivery were included. Fetuses with known structural or genetic abnormalities were excluded. Pearson's correlation coefficients were determined for both 2D and 3D EFW to BW then compared using Williams' test and Fisher r to z transformation, where applicable. Mean percent difference and standard deviation were used to assess the accuracy and precision, respectively, of 2D and 3D EFWs in BW prediction. RESULTS Two hundred forty-eight pregnancies were included. Ultrasound studies were performed with a median interval of 2 weeks (IQR 1, 3) between ultrasound and delivery. Both 2D and 3D estimated fetal weights showed a significant correlation with birth weight (r = 0.74 and r = 0.73, respectively), indicating similar accuracy between the two techniques. CONCLUSION Two-dimensional and three-dimensional EFWs performed similarly in the prediction of BW in borderline small fetuses.
Collapse
Affiliation(s)
- Felicia V LeMoine
- The MetroHealth System, Case Western Reserve University Program, Cleveland, Ohio, USA
| | - Hannah Hill
- The MetroHealth System, Population Health and Equity Research Institute, Cleveland, Ohio, USA
| | - Marisa R Imbroane
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Aishwarya A Gautam
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chloe H Van Dorn
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Angela C Ranzini
- The MetroHealth System, Department of Reproductive Biology, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Watt GE, Hubbard ME, Mottola MF. The association of physical activity during pregnancy with birthweight extremes: A scoping review. J Sci Med Sport 2024; 27:684-690. [PMID: 38866653 DOI: 10.1016/j.jsams.2024.05.016] [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: 11/14/2023] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVES Birthweight (BW) indicates newborn health and is a risk factor for development of chronic diseases later in life. The aim was to investigate whether or not prenatal physical activity (PA) influences BW extremes and how PA influences BW extremes in those diagnosed with pregnancy-related diseases. DESIGN We performed a scoping review. METHODS Searches were completed on five databases and studies identified were uploaded to Covidence. RESULTS Across the five databases 3114 studies were identified and after screening, 69 of these studies were used for the final review. Of the 61 studies that considered low BW (LBW)/small for gestational age (SGA) infants, the majority of results (69 %) indicated that PA during pregnancy had no significant impact on LBW or SGA infants. In addition, 11 % of studies reported a significant decrease in the prevalence of LBW infants, however two studies (3 %) reported a significant increase in LBW or SGA infants, likely relating to individuals with high body mass index and poor adherence to PA. Of the 41 studies that did report LGA/macrosomia, 34 % reported that PA significantly reduced the prevalence of higher BW infants. One study reported the association between meeting exercise recommendations and reducing the odds of LGA infants in those with pregnancy-related diseases. CONCLUSIONS We provide evidence on the association of prenatal PA with BW extremes. It is suggested that prenatal PA does not increase the risk of delivering LBW/SGA infants and may reduce the prevalence of large BW infants. Further research is needed to confirm these relationships and explain their underlying mechanisms.
Collapse
Affiliation(s)
- Grace Emma Watt
- R. Samuel McLaughlin Foundation - Exercise and Pregnancy Lab, University of Western Ontario, Canada; School of Kinesiology, Faculty of Health Sciences, University of Western Ontario, Canada
| | - Madeline E Hubbard
- R. Samuel McLaughlin Foundation - Exercise and Pregnancy Lab, University of Western Ontario, Canada; School of Kinesiology, Faculty of Health Sciences, University of Western Ontario, Canada; Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Michelle F Mottola
- R. Samuel McLaughlin Foundation - Exercise and Pregnancy Lab, University of Western Ontario, Canada; School of Kinesiology, Faculty of Health Sciences, University of Western Ontario, Canada; Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada; Children's Health Research Institute, University of Western Ontario, Canada.
| |
Collapse
|
9
|
Bertossa MR, Darby JR, Holman SL, Meakin AS, Li C, Huber HF, Wiese MD, Nathanielsz PW, Morrison JL. Maternal high fat-high energy diet alters metabolic factors in the non-human primate fetal heart. J Physiol 2024; 602:4251-4269. [PMID: 39087821 PMCID: PMC11366491 DOI: 10.1113/jp286861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
The consumption of high fat-high energy diets (HF-HEDs) continues to rise worldwide and parallels the rise in maternal obesity (MO) that predisposes offspring to cardiometabolic disorders. Although the underlying mechanisms are unclear, thyroid hormones (TH) modulate cardiac maturation in utero. Therefore, we aimed to determine the impact of a high fat-high energy diet (HF-HED) on the hormonal, metabolic and contractility profile of the non-human primate (NHP) fetal heart. At ∼9 months preconception, female baboons (Papio hamadryas) were randomly assigned to either a control diet or HF-HED. At 165 days gestational age (term = 184 days), fetuses were delivered by Caesarean section under anaesthesia, humanely killed, and left ventricular cardiac tissue (Control (n = 6 female, 6 male); HF-HED (n = 6 F, 6 M)) was collected. Maternal HF-HED decreased the concentration of active cardiac TH (i.e. triiodothyronine (T3)), and type 1 iodothyronine deiodinase (DIO1) mRNA expression. Maternal HF-HED decreased the abundance of cardiac markers of insulin-mediated glucose uptake phosphorylated insulin receptor substrate 1 (Ser789) and glucose transporter 4, and increased protein abundance of key oxidative phosphorylation complexes (I, III, IV) and mitochondrial abundance in both sexes. Maternal HF-HED alters cardiac TH status, which may induce early signs of cardiac insulin resistance. This may increase the risk of cardiometabolic disorders in later life in offspring born to these pregnancies. KEY POINTS: Babies born to mothers who consume a high fat-high energy diet (HF-HED) prior to and during pregnancy are predisposed to an increased risk of cardiometabolic disorders across the life course. Maternal HF-HED prior to and during pregnancy decreased thyroid hormone triiodothyronine (T3) concentrations and type 1 iodothyronine deiodinase DIO1 mRNA expression in the non-human primate fetal heart. Maternal HF-HED decreased markers of insulin-dependent glucose uptake, phosphorylated insulin receptor substrate 1 and glucose transporter 4 in the fetal heart. Maternal HF-HED increased mitochondrial abundance and mitochondrial OXPHOS complex I, III and IV in the fetal heart. Fetuses from HF-HED pregnancies are predisposed to cardiometabolic disorders that may be mediated by changes in T3, placing them on a poor lifetime cardiovascular health trajectory.
Collapse
Affiliation(s)
- Melanie R. Bertossa
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation
| | - Jack R.T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation
| | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Hillary F. Huber
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia, 5001
| | | | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation
| |
Collapse
|
10
|
Hao X, Long X, Fan L, Gou J, Liu Y, Fu Y, Zhao H, Xie X, Wang D, Liang G, Ye Y, Wang J, Li S, Zeng C. Prenatal LPS leads to increases in RAS expression within the PVN and overactivation of sympathetic outflow in offspring rats. Hypertens Res 2024; 47:2363-2376. [PMID: 38969805 PMCID: PMC11374713 DOI: 10.1038/s41440-024-01754-z] [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: 12/02/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 07/07/2024]
Abstract
The renin-angiotensin system (RAS) and the sympathetic nervous system (SNS) are two major blood pressure-regulating systems. The link between the renal and cerebral RAS axes was provided by reflex activation of renal afferents and efferent sympathetic nerves. There is a self-sustaining enhancement of the brain and the intrarenal RAS. In this study, prenatal exposure to lipopolysaccharide (LPS) led to increased RAS activity in the paraventricular nucleus (PVN) and overactivation of sympathetic outflow, accompanied by increased production of reactive oxygen species (ROS) and disturbances between inhibitory and excitatory neurons in PVN. The AT1 receptor blocker losartan and α2 adrenergic receptor agonist clonidine in the PVN significantly decreased renal sympathetic nerve activity (RSNA) and synchronously reduced systolic blood pressure. Prenatal LPS stimulation caused H3 acetylation at H3K9 and H3K14 in the PVN, which suggested that epigenetic changes are involved in transmitting the prenatal adverse stimulative information to the next generation. Additionally, melatonin treatment during pregnancy reduced RAS activity and ROS levels in the PVN; balanced the activity of inhibitory and excitatory neurons in the PVN; increased urine sodium secretion; reduced RSNA and blood pressure. In conclusion, prenatal LPS leads to increased RAS expression within the PVN and overactivation of the sympathetic outflow, thereby contributing to hypertension in offspring rats. Melatonin is expected to be a promising agent for preventing prenatal LPS exposure-induced hypertension.
Collapse
Affiliation(s)
- Xueqin Hao
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Department of Anesthesiology, the First affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Xueting Long
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Lingling Fan
- Department of Physiology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jijia Gou
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yuchao Liu
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yifan Fu
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Huijuan Zhao
- Department of human Anatomy and Histoembryology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xiaojuan Xie
- Department of Anesthesiology, the First affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Dongmei Wang
- Department of Microbiology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Gaofeng Liang
- Department of Pathology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yujia Ye
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Jing Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Sanqiang Li
- Department of Biochemistry, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
11
|
Ho SY, Yuliana ME, Chou HC, Huang LT, Chen CM. Altered purine and pentose phosphate pathway metabolism in uteroplacental insufficiency-induced intrauterine growth restriction offspring rats impair intestinal function. J Nutr Biochem 2024; 134:109737. [PMID: 39147244 DOI: 10.1016/j.jnutbio.2024.109737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
This study aimed to identify metabolic alterations in the small intestine of newborn rats with intrauterine growth restriction (IUGR), a condition linked to intestinal dysfunction. Pregnant Sprague Dawley rats underwent bilateral uterine artery ligation on gestational day 17 to induce intrauterine growth restriction or sham surgery. Rat pups were delivered spontaneously on gestational day 22. Small intestine tissues were collected on postnatal days 0 and 7 from offspring. Liquid chromatography-mass spectrometry analysis was performed to investigate untargeted metabolomic profiles. Western blot analysis assessed protein expression of key regulators. Newborn rats with intrauterine growth restriction exhibited distinct small intestine metabolic profiles compared to controls on postnatal day 0. Notably, significant alterations were observed in purine metabolism, the pentose phosphate pathway, and related pathways. Western blot analysis revealed a decrease expression in transketolase, a key enzyme of the pentose phosphate pathway, suggesting impaired activity of the pentose phosphate pathway. Additionally, decreased expression of tight junction proteins ZO-1 and occludin indicated compromised intestinal barrier function in rats with intrauterine growth restriction. Similar metabolic disruptions persisted on postnatal day 7, with further reductions in tricarboxylic acid cycle intermediates and folate biosynthesis precursors. Interestingly, lysyl-glycine, a protein synthesis marker, was elevated in rats with intrauterine growth restriction. Our findings reveal a distinct metabolic signature in the small intestine of neonatal rats with intrauterine growth restriction, characterized by disruptions in the pentose phosphate pathway, purine metabolism, and energy production pathways. These novel insights suggest potential mechanisms underlying IUGR-associated intestinal dysfunction and impaired growth.
Collapse
Affiliation(s)
- Sheng-Yuan Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Merryl Esther Yuliana
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Faculty of Medicine, Christian University of Indonesia, Jakarta, Indonesia
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Liang-Ti Huang
- Department of Pediatrics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ming Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
12
|
Yanik T, Katirci E, Simsek M, Korgun ET, Kipmen-Korgun D. Effects of Hyperglycemia on Angiogenesis in Human Placental Endothelial Cells. Z Geburtshilfe Neonatol 2024; 228:346-354. [PMID: 38740370 DOI: 10.1055/a-2282-9007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The placenta is a temporary organ that provides communication between the mother and fetus. Maternal diabetes and abnormal placental angiogenesis may be linked. We investigated the angiogenesis mechanism resulting from VEGF and glucose stimulation in PECs obtained from human term placenta. Immunohistochemistry was performed to characterize PECs obtained from human term placenta. D-glucose was added to the medium containing PECs to establish normoglycemic and hyperglycemic conditions. The expression levels of VEGF, VEGFR-1 and VEGFR-2 genes and proteins in PECs from the control and experimental groups were analyzed by RT-PCR and Western blotting, respectively. With 48-hours incubation, gene expressions increased due to hyperglycemia, while protein levels increased due to the combined effect of VEGF and hyperglycemia. While VEGFR-2 gene expression and protein amounts increased in 24-hours due to the combined effect of VEGF and hyperglycemia, the effect of VEGF stimulation and glucose level on VEGFR-2 decreased in 48-hour incubation with time. VEGF, VEGFR-1 and VEGFR-2 genes and proteins were affected by hyperglycemic conditions in PECs. Hyperglycemia occurring in various conditions such as gestational diabetes mellitus and diabetes mellitus may affect VEGF, VEGFR-1 and VEGFR-2 genes and proteins of PECs derived from human term placenta.
Collapse
Affiliation(s)
- Turkan Yanik
- Histology and Embryology, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Ertan Katirci
- Histology and Embryology, Ahi Evran University Faculty of Medicine, Kirsehir, Turkey
| | - Mehmet Simsek
- Obstetrics And Gynaecology, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Emin Turkay Korgun
- Histology and Embryology, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Dijle Kipmen-Korgun
- Department Of Medical Biochemistry, Akdeniz University Faculty of Medicine, Antalya, Turkey
| |
Collapse
|
13
|
Bozdemir N, Kablan T, Altintas MO, Sukur G, Cinar O, Uysal F. Altered DNA methylation and Dnmt expression in obese uterus may cause implantation failure. J Mol Histol 2024; 55:427-436. [PMID: 38850446 DOI: 10.1007/s10735-024-10212-6] [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: 10/28/2023] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Obesity is defined by increased adipose tissue volume and has become a major risk factor for reproduction. Recent studies have revealed a substantial link between obesity and epigenetics. The epigenome is dynamically regulated mainly by DNA methylation. DNA methylation, which is controlled by DNA methyltransferases (Dnmts), has been widely studied because it is essential for imprinting and regulation of gene expression. In our previous study, we showed that the levels of Dnmt1, Dnmt3a and global DNA methylation was dramatically altered in the testis and ovary of high-fat diet (HFD)-induced obese mice. However, the effect of HFD on Dnmts and global DNA methylation in mouse uterus has not yet been demonstrated. Therefore, in the present study, we aimed to evaluate the effect of HFD on the level of Dnmt1, Dnmt3a, Dnmt3b, Dnmt3l and global DNA methylation in uterus. Our results showed that HFD significantly altered the levels of Dnmts and global DNA methylation in the uterus. The total expression of Dnmt1, Dnmt3a and Dnmt3b was significantly upregulated, while level of Dnmt3l and global DNA methylation were dramatically decreased (p < 0.05). Furthermore, we observed that the expression of Dnmt3b and Dnmt3l was significantly increased in endometrium including gland and epithelium (p < 0.05). Although Dnmt3b was the only protein whose expression significantly increased, the level of global DNA methylation and Dnmt3l significantly decreased in stroma and myometrium (p < 0.05). In conclusion, our results show for the first time that obesity dramatically alters global DNA methylation and expression of Dnmts, and decreased DNA methylation and Dnmt expression may cause abnormal gene expression, especially in the endometrium.
Collapse
Affiliation(s)
- Nazlican Bozdemir
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey
| | - Tuba Kablan
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey
| | - Mehmet Ozgen Altintas
- Department of Physiology, Ankara Medipol University School of Medicine, Ankara, Turkey
- Department of Physiology, Istanbul Medipol University Institute of Health Sciences, Istanbul, Turkey
| | - Gozde Sukur
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Fatma Uysal
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey.
| |
Collapse
|
14
|
Zhang S, Lock MC, Tie M, McMillen IC, Botting KJ, Morrison JL. Cardiac programming in the placentally restricted sheep fetus in early gestation. J Physiol 2024; 602:3815-3832. [PMID: 38975864 DOI: 10.1113/jp286702] [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: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
Fetal growth restriction (FGR) occurs in 8% of human pregnancies, and the growth restricted newborn is at a greater risk of developing heart disease in later adult life. In sheep, experimental restriction of placental growth (PR) from conception results in FGR, a decrease in cardiomyocyte endowment and an upregulation of pathological hypertrophic signalling in the fetal heart in late gestation. However, there is no change in the expression of markers of cellular proliferation nor in the level of cardiomyocyte apoptosis in the heart of the PR fetus in late gestation. This suggests that FGR arises early in gestation and programs a decrease in cardiomyocyte endowment in early, rather than late, gestation. Here, control and PR fetal sheep were humanely killed at 55 days' gestation (term, 150 days). Fetal body and heart weight were lower in PR compared with control fetuses and there was evidence of sparing of fetal brain growth. While there was no change in the proportion of cardiomyocytes that were proliferating in the early gestation PR heart, there was an increase in measures of apoptosis, and markers of autophagy and pathological hypertrophy in the PR fetal heart. These changes in early gestation highlight that FGR is associated with evidence of early cell death and compensatory hypertrophic responses of cardiomyocytes in the fetal heart. The data suggest that early placental restriction results in a decrease in the pool of proliferative cardiomyocytes in early gestation, which would limit cardiomyocyte endowment in the heart of the PR fetus in late gestation. KEY POINTS: Placental restriction leading to fetal growth restriction (FGR) and chronic fetal hypoxaemia in sheep results in a decrease in cardiomyocyte endowment in late gestation. FGR did not change cardiomyocyte proliferation during early gestation but did result in increased apoptosis and markers of autophagy in the fetal heart, which may result in the decreased endowment of cardiomyocytes observed in late gestation. FGR in early gestation also results in increased hypoxia inducible factor signalling in the fetal heart, which in turn may result in the altered expression of epigenetic regulators, increased expression of insulin-like growth factor 2 and cardiomyocyte hypertrophy during late gestation and after birth.
Collapse
Affiliation(s)
- Song Zhang
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Michelle Tie
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
15
|
dos Santos FCR, Santarosa BP, Dal Más FE, da Silva KN, Guirro ÉCBDP, Gomes V. Effects of dam metabolic profile and seasonality (Spring vs. Winter) on their offspring' metabolism, health, and immunity: maternal factors in dairy calves' analytes. Front Vet Sci 2024; 11:1424960. [PMID: 39076303 PMCID: PMC11285391 DOI: 10.3389/fvets.2024.1424960] [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: 05/13/2024] [Accepted: 06/14/2024] [Indexed: 07/31/2024] Open
Abstract
Maternal status during the transition period can significantly impact the health and performance of Holstein dairy calves, with lasting effects on various variables. However, the relationship between maternal late gestation metabolic status, seasonality, and their impact on offspring remains unclear. This study aimed to assess the influence of maternal variables at calving on the performance, metabolism, and immunity of 28 dairy calves during their first month of life. Blood samples were collected from 28 Holstein cows at calving. Median results for maternal variables including non-esterified fatty acids (NEFA), β-hydroxybutyrate (BHB), glucose, total protein (TP), albumin, triglycerides (TG), total cholesterol (TC), haptoglobin (Hp), body weight (BW), and body condition score (BCS) were determined. These median values served as a basis for categorizing the offspring into two groups based on their dams' high or low degree of each maternal variable. Additionally, calves were categorized by the season of birth (Spring vs. Winter), with 14 in each. Blood samples were collected from the calves at birth and on days 1, 7, 14, and 28 to assess IgG, biochemical parameters, and haptoglobin concentration. Reactive oxygen species (ROS) production by polymorphonuclear cells stimulated by various agents was also evaluated. Clinical assessments were conducted for diarrhea and bovine respiratory disease frequencies. Despite the overall health of the cows, differences were observed in the calves between maternal groups. Heavier cows with high maternal BCS tended to have larger offspring, while high maternal BCS was associated with increased diarrhea prevalence. Low maternal BCS resulted in a stronger innate immune response, indicated by higher ROS production. Calves from cows experiencing metabolic changes during calving displayed elevated Hp concentrations. Spring-born calves were larger but had lower serum IgG concentration and reduced innate immune response compared to winter-born calves. Additionally, spring-born calves exhibited higher Hp and increased diarrhea prevalence on day 28. These findings underscore the importance of the prenatal period in determining neonatal health and suggest further research to elucidate the long-term clinical implications of maternal effects on offspring health and growth. Investigating offspring constituents later in life can provide insight into the persistence of maternal effects over time.
Collapse
Affiliation(s)
| | - Bianca Paola Santarosa
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Felipe Eduardo Dal Más
- Departament of Veterinary Science, Federal University of Paraná, Palotina, Paraná State, Brazil
| | - Karen Nascimento da Silva
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Viviani Gomes
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Jiang X, Yang G, Feng N, Du X, Xu L, Zhong VW. Lifestyle modifies the associations of early-life smoking behaviors and genetic susceptibility with type 2 diabetes: A prospective cohort study involving 433,872 individuals from UK Biobank. Diabetes Metab Syndr 2024; 18:103090. [PMID: 39084054 DOI: 10.1016/j.dsx.2024.103090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND To investigate whether and what lifestyle factors in later life modify the associations of early-life smoking behaviors and genetic susceptibility with type 2 diabetes (T2D). METHODS In the UK Biobank, in utero tobacco exposure (n = 354,493) and age of smoking initiation (n = 353,557) were self-reported. A composite lifestyle score was calculated based on diet, physical activity, nicotine exposure, sleep duration, and BMI. Hazard ratio (HR) and absolute risk difference (ARD) were used to estimate the associations of early-life smoking behaviors and genetic risk with incident T2D, as well as the effect modification of the lifestyle score. RESULTS During a median follow-up of 14.6 years, the HRs (95 % CIs) of T2D for in utero tobacco exposure, and smoking initiation in adulthood, adolescence, and childhood, compared with no smoking behavior, were 1.19 (1.16-1.23), 1.34 (1.29-1.39), 1.58 (1.53-1.64), 2.22 (2.11-2.32), respectively (P for trend<0.001). Early-life smoking behaviors and high genetic risk (vs no smoking behavior and low genetic risk) were associated with a 302%-593 % higher T2D risk (P for additive interaction<0.05). Compared to participants with early-life smoking behaviors, high genetic risk, and an unfavorable lifestyle, those who adhered to a favorable lifestyle had a lower T2D risk in all subgroups (HRs from 0.05 to 0.36 and ARD from -14.97 % to -9.51 %), with the highest ARD attributable to lifestyle in participants with early-life smoking behaviors and high genetic risk. CONCLUSIONS The T2D risk associated with early-life smoking behaviors and genetic risk was modified by a favorable lifestyle.
Collapse
Affiliation(s)
- Xuanwei Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangrui Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nannan Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xihao Du
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Victor W Zhong
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
17
|
Darby JRT, Saini BS, Holman SL, Hammond SJ, Perumal SR, Macgowan CK, Seed M, Morrison JL. Acute-on-chronic: using magnetic resonance imaging to disentangle the haemodynamic responses to acute and chronic fetal hypoxaemia. Front Med (Lausanne) 2024; 11:1340012. [PMID: 38933113 PMCID: PMC11199546 DOI: 10.3389/fmed.2024.1340012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction The fetal haemodynamic response to acute episodes of hypoxaemia are well characterised. However, how these responses change when the hypoxaemia becomes more chronic in nature such as that associated with fetal growth restriction (FGR), is less well understood. Herein, we utilised a combination of clinically relevant MRI techniques to comprehensively characterize and differentiate the haemodynamic responses occurring during acute and chronic periods of fetal hypoxaemia. Methods Prior to conception, carunclectomy surgery was performed on non-pregnant ewes to induce FGR. At 108-110 days (d) gestational age (GA), pregnant ewes bearing control (n = 12) and FGR (n = 9) fetuses underwent fetal catheterisation surgery. At 117-119 days GA, ewes underwent MRI sessions where phase-contrast (PC) and T2 oximetry were used to measure blood flow and oxygenation, respectively, throughout the fetal circulation during a normoxia and then an acute hypoxia state. Results Fetal oxygen delivery (DO2) was lower in FGR fetuses than controls during the normoxia state but cerebral DO2 remained similar between fetal groups. Acute hypoxia reduced both overall fetal and cerebral DO2. FGR increased ductus venosus (DV) and foramen ovale (FO) blood flow during both the normoxia and acute hypoxia states. Pulmonary blood flow (PBF) was lower in FGR fetuses during the normoxia state but similar to controls during the acute hypoxia state when PBF in controls was decreased. Conclusion Despite a prevailing level of chronic hypoxaemia, the FGR fetus upregulates the preferential streaming of oxygen-rich blood via the DV-FO pathway to maintain cerebral DO2. However, this upregulation is unable to maintain cerebral DO2 during further exposure to an acute episode of hypoxaemia. The haemodynamic alterations required at the level of the liver and lung to allow the DV-FO pathway to maintain cerebral DO2, may have lasting consequences on hepatic function and pulmonary vascular regulation after birth.
Collapse
Affiliation(s)
- Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Brahmdeep S. Saini
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sarah J. Hammond
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sunthara Rajan Perumal
- Preclinical, Imaging & Research Laboratories, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Christopher K. Macgowan
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
| | - Mike Seed
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Deng WQ, Pigeyre M, Azab SM, Wilson SL, Campbell N, Cawte N, Morrison KM, Atkinson SA, Subbarao P, Turvey SE, Moraes TJ, Mandhane P, Azad MB, Simons E, Pare G, Anand SS. Consistent cord blood DNA methylation signatures of gestational age between South Asian and white European cohorts. Clin Epigenetics 2024; 16:74. [PMID: 38840168 PMCID: PMC11155053 DOI: 10.1186/s13148-024-01684-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Epigenetic modifications, particularly DNA methylation (DNAm) in cord blood, are an important biological marker of how external exposures during gestation can influence the in-utero environment and subsequent offspring development. Despite the recognized importance of DNAm during gestation, comparative studies to determine the consistency of these epigenetic signals across different ethnic groups are largely absent. To address this gap, we first performed epigenome-wide association studies (EWAS) of gestational age (GA) using newborn cord blood DNAm comparatively in a white European (n = 342) and a South Asian (n = 490) birth cohort living in Canada. Then, we capitalized on established cord blood epigenetic GA clocks to examine the associations between maternal exposures, offspring characteristics and epigenetic GA, as well as GA acceleration, defined as the residual difference between epigenetic and chronological GA at birth. RESULTS Individual EWASs confirmed 1,211 and 1,543 differentially methylated CpGs previously reported to be associated with GA, in white European and South Asian cohorts, respectively, with a similar distribution of effects. We confirmed that Bohlin's cord blood GA clock was robustly correlated with GA in white Europeans (r = 0.71; p = 6.0 × 10-54) and South Asians (r = 0.66; p = 6.9 × 10-64). In both cohorts, Bohlin's clock was positively associated with newborn weight and length and negatively associated with parity, newborn female sex, and gestational diabetes. Exclusive to South Asians, the GA clock was positively associated with the newborn ponderal index, while pre-pregnancy weight and gestational weight gain were strongly predictive of increased epigenetic GA in white Europeans. Important predictors of GA acceleration included gestational diabetes mellitus, newborn sex, and parity in both cohorts. CONCLUSIONS These results demonstrate the consistent DNAm signatures of GA and the utility of Bohlin's GA clock across the two populations. Although the overall pattern of DNAm is similar, its connections with the mother's environment and the baby's anthropometrics can differ between the two groups. Further research is needed to understand these unique relationships.
Collapse
Affiliation(s)
- Wei Q Deng
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Canada.
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada.
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada.
| | - Marie Pigeyre
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
| | - Sandi M Azab
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Samantha L Wilson
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Canada
| | - Natalie Campbell
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Nathan Cawte
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada
| | | | | | - Padmaja Subbarao
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
- Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
- Program in Translational Medicine, SickKids Research Institute, Toronto, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, Canada
| | - Theo J Moraes
- Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
- Program in Translational Medicine, SickKids Research Institute, Toronto, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Elinor Simons
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Guillaume Pare
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Sonia S Anand
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada.
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada.
| |
Collapse
|
19
|
Ghasemi Z, Alizadeh Mogadam Masouleh A, Rashki Ghaleno L, Akbarinejad V, Rezazadeh Valojerdi M, Shahverdi A. Maternal nutrition and fetal imprinting of the male progeny. Anim Reprod Sci 2024; 265:107470. [PMID: 38657462 DOI: 10.1016/j.anireprosci.2024.107470] [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: 12/28/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/26/2024]
Abstract
The global population as well as the demand for human food is rapidly growing worldwide, which necessitates improvement of efficiency in livestock operations. In this context, environmental factors during fetal and/or neonatal life have been observed to influence normal physical and physiological function of an individual during adulthood, and this phenomenon is called fetal or developmental programming. While numerous studies have reported the impact of maternal factors on development of the female progeny, limited information is available on the potential effects of fetal programming on reproductive function of the male offspring. Therefore, the objective for this review article was to focus on available literature regarding the impact of maternal factors, particularly maternal nutrition, on reproductive system of the male offspring. To this end, we highlighted developmental programming of the male offspring in domestic species (i.e., pig, cow and sheep) as well as laboratory species (i.e., mice and rat) during pregnancy and lactation. In this sense, we pointed out the effects of maternal nutrition on various functions of the male offspring including hypothalamic-pituitary axis, hormonal levels, testicular tissue and semen parameters.
Collapse
Affiliation(s)
- Zahrasadat Ghasemi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - AliReza Alizadeh Mogadam Masouleh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Gyn-medicum, Center for Reproductive Medicine, Göttingen, Germany; Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.
| | - Leila Rashki Ghaleno
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
20
|
Akyüz B, Sohel MMH, Konca Y, Arslan K, Gürbulak K, Abay M, Kaliber M, White SN, Cinar MU. Effects of Low and High Maternal Protein Intake on Fetal Skeletal Muscle miRNAome in Sheep. Animals (Basel) 2024; 14:1594. [PMID: 38891641 PMCID: PMC11171157 DOI: 10.3390/ani14111594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Prenatal maternal feeding plays an important role in fetal development and has the potential to induce long-lasting epigenetic modifications. MicroRNAs (miRNAs) are non-coding, single-stranded RNAs that serve as one epigenetic mechanism. Though miRNAs have crucial roles in fetal programming, growth, and development, there is limited data regarding the maternal diet and miRNA expression in sheep. Therefore, we analyzed high and low maternal dietary protein for miRNA expression in fetal longissimus dorsi. Pregnant ewes were fed an isoenergetic high-protein (HP, 160-270 g/day), low-protein (LP, 73-112 g/day), or standard-protein diet (SP, 119-198 g/day) during pregnancy. miRNA expression profiles were evaluated using the Affymetrix GeneChip miRNA 4.0 Array. Twelve up-regulated, differentially expressed miRNAs (DE miRNAs) were identified which are targeting 65 genes. The oar-3957-5p miRNA was highly up-regulated in the LP and SP compared to the HP. Previous transcriptome analysis identified that integrin and non-receptor protein tyrosine phosphatase genes targeted by miRNAs were detected in the current experiment. A total of 28 GO terms and 10 pathway-based gene sets were significantly (padj < 0.05) enriched in the target genes. Most genes targeted by the identified miRNAs are involved in immune and muscle disease pathways. Our study demonstrated that dietary protein intake during pregnancy affected fetal skeletal muscle epigenetics via miRNA expression.
Collapse
Affiliation(s)
- Bilal Akyüz
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
| | - Md Mahmodul Hasan Sohel
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
- Genome and Stem Cell Centre, Erciyes University, Kayseri 38039, Türkiye
| | - Yusuf Konca
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
| | - Korhan Arslan
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
| | - Kutlay Gürbulak
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (K.G.); (M.A.)
| | - Murat Abay
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (K.G.); (M.A.)
| | - Mahmut Kaliber
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
| | - Stephen N. White
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA 99164, USA;
| | - Mehmet Ulas Cinar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
21
|
Mobedi E, Harati HRD, Allahyari I, Gharagozlou F, Vojgani M, Baghbanani RH, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: IV. Association of maternal milk fat and protein percentage and milk fat to protein ratio with offspring's birth weight, survival, productive and reproductive performance and AMH concentration from birth to the first lactation period. Theriogenology 2024; 220:12-25. [PMID: 38457855 DOI: 10.1016/j.theriogenology.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
Although the association of maternal milk production with developmental programming of offspring has been investigated, there is limited information available on the relationship of maternal milk components with productive and reproductive performance of the offspring. Therefore, the present study was conducted to analyze the association of maternal milk fat and protein percentage and milk fat to protein ratio with birth weight, survival, productive and reproductive performance and AMH concentration in the offspring. In study I, data of birth weight, milk yield and reproductive variables of offspring born to lactating dams (n = 14,582) and data associated with average maternal milk fat percentage (MFP), protein percentage (MPP) and fat to protein ratio (MFPR) during 305-day lactation were retrieved. Afterwards, offspring were classified in various categories of MFP, MPP and MFPR. In study II, blood samples (n = 339) were collected from offspring in various categories of MFP, MPP and MFPR for measurement of serum AMH. Maternal milk fat percentage was positively associated with birth weight and average percentage of milk fat (APMF) and protein (APMP) and milk fat to protein ratio (FPR) during the first lactation, but negatively associated with culling rate during nulliparity in the offspring (P < 0.05). Maternal milk protein percentage was positively associated with birth weight, APMF, APMP, FPR and culling rate, but negatively associated with milk yield and fertility in the offspring (P < 0.05). Maternal FPR was positively associated with APMF and FPR, but negatively associated with culling rate, APMP and fertility in the offspring (P < 0.05). However, concentration of AMH in the offspring was not associated with MFP, MPP and MFPR (P > 0.05). In conclusion, the present study revealed that maternal milk fat and protein percentage and their ratio were associated with birth weight, survival, production and reproduction of the offspring. Yet it was a preliminary research and further studies are required to elucidate the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
22
|
Akbarinejad V, Cushman RA. Developmental programming of reproduction in the female animal. Anim Reprod Sci 2024; 263:107456. [PMID: 38503204 DOI: 10.1016/j.anireprosci.2024.107456] [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: 12/24/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Successful reproduction is a cornerstone in food animal industry in order to sustain food production for human. Therefore, various methods focusing on genetics and postnatal environment have been identified and applied to improve fertility in livestock. Yet there is evidence indicating that environmental factors during prenatal and/or neonatal life can also impact the function of reproductive system and fertility in the animals during adulthood, which is called the developmental programming of reproduction. The current review summarizes data associated with the developmental origins of reproduction in the female animals. In this regard, this review focuses on the effect of plane of nutrition, maternal body condition, hypoxia, litter size, maternal age, parity, level of milk production and milk components, lactocrine signaling, stress, thermal stress, exposure to androgens, endocrine disrupting chemicals, mycotoxins and pollutants, affliction with infection and inflammation, and maternal gut microbiota during prenatal and neonatal periods on the neuroendocrine system, puberty, health of reproductive organs and fertility in the female offspring. It is noteworthy that these prenatal and neonatal factors do not always exert their effects on the reproductive performance of the female by compromising the development of organs directly related to reproductive function such as hypothalamus, pituitary, ovary, oviduct and uterus. Since they can impair the development of non-reproductive organs and systems modulating reproductive function as well (e.g., metabolic system and level of milk yield in dairy animals). Furthermore, when these factors affect the epigenetics of the offspring, their adverse effects will not be limited to one generation and can transfer transgenerationally. Hence, pinpointing the factors influencing developmental programming of reproduction and considering them in management of livestock operations could be a potential strategy to help improve fertility in food animals.
Collapse
Affiliation(s)
- Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Robert A Cushman
- USDA, Agricultural Research Service, US. Meat Animal Research Center, Clay Center, NE 68933-0166, United States
| |
Collapse
|
23
|
Schroeder M, Fuenzalida B, Yi N, Shahnawaz S, Gertsch J, Pellegata D, Ontsouka E, Leiva A, Gutiérrez J, Müller M, Brocco MA, Albrecht C. LAT1-dependent placental methionine uptake is a key player in fetal programming of metabolic disease. Metabolism 2024; 153:155793. [PMID: 38295946 DOI: 10.1016/j.metabol.2024.155793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
The Developmental Origins of Health and Disease hypothesis sustains that exposure to different stressors during prenatal development prepares the offspring for the challenges to be encountered after birth. We studied the gestational period as a particularly vulnerable window where different stressors can have strong implications for fetal programming of the offspring's life-long metabolic status via alterations of specific placentally expressed nutrient transporters. To study this mechanism, we used a murine prenatal stress model, human preeclampsia, early miscarriage, and healthy placental tissue samples, in addition to in vitro models of placental cells. In stressed mice, placental overexpression of L-type amino acid transporter 1 (Lat1) and subsequent global placental DNA hypermethylation was accompanied by fetal and adult hypothalamic dysregulation in global DNA methylation and gene expression as well as long-term metabolic abnormalities exclusively in female offspring. In human preeclampsia, early miscarriage, and under hypoxic conditions, placental LAT1 was significantly upregulated, leading to increased methionine uptake and global DNA hypermethylation. Remarkably, subgroups of healthy term placentas with high expression of stress-related genes presented increased levels of placental LAT1 mRNA and protein, DNA and RNA hypermethylation, increased methionine uptake capacity, one-carbon metabolic pathway disruption, higher methionine concentration in the placenta and transport to the fetus specifically in females. Since LAT1 mediates the intracellular accumulation of methionine, global DNA methylation, and one-carbon metabolism in the placenta, our findings hint at a major sex-specific global response to a variety of prenatal stressors affecting placental function, epigenetic programming, and life-long metabolic disease and provide a much-needed insight into early-life factors predisposing females/women to metabolic disorders.
Collapse
Affiliation(s)
- Mariana Schroeder
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| | - Barbara Fuenzalida
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Nan Yi
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Saira Shahnawaz
- Department of Biochemistry, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan; Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan
| | - Jürg Gertsch
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Daniele Pellegata
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Edgar Ontsouka
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Andrea Leiva
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Jaime Gutiérrez
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Martin Müller
- Division of Gynecology and Obstetrics, Lindenhofgruppe, Bern, Switzerland
| | - Marcela A Brocco
- Institute of Biotechnological Research, University of San Martín, Buenos Aires, Argentina
| | - Christiane Albrecht
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Ruuskanen S. Early-life environmental effects on birds: epigenetics and microbiome as mechanisms underlying long-lasting phenotypic changes. J Exp Biol 2024; 227:jeb246024. [PMID: 38449325 DOI: 10.1242/jeb.246024] [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
Although the long-lasting effects of variation in early-life environment have been well documented across organisms, the underlying causal mechanisms are only recently starting to be unraveled. Yet understanding the underlying mechanisms of long-lasting effects can help us predict how organisms will respond to changing environments. Birds offer a great system in which to study developmental plasticity and its underlying mechanisms owing to the production of large external eggs and variation in developmental trajectories, combined with a long tradition of applied, physiological, ecological and evolutionary research. Epigenetic changes (such as DNA methylation) have been suggested to be a key mechanism mediating long-lasting effects of the early-life environment across taxa. More recently, changes in the early-life gut microbiome have been identified as another potential mediator of developmental plasticity. As a first step in understanding whether these mechanisms contribute to developmental plasticity in birds, this Review summarizes how changes in early-life environment (both prenatal and postnatal) influence epigenetic markers and the gut microbiome. The literature shows how both early-life biotic (such as resources and social environment) and abiotic (thermal environment and various anthropogenic stressors) factors modify epigenetic markers and the gut microbiome in birds, yet data concerning many other environmental factors are limited. The causal links of these modifications to lasting phenotypic changes are still scarce, but changes in the hypothalamic-pituitary-adrenal axis have been identified as one putative pathway. This Review identifies several knowledge gaps, including data on the long-term effects, stability of the molecular changes, and lack of diversity in the systems studied, and provides directions for future research.
Collapse
Affiliation(s)
- Suvi Ruuskanen
- Department of Biological and Environmental Science, University of Jyväskylä, Survontie 9C, 40500 Jyväskylä, Finland
- Department of Biology, University of Turku, Vesilinnankatu 5, 20500 Turku, Finland
| |
Collapse
|
25
|
Olga L, McKenzie K, Kerac M, Boyne M, Badaloo A, Bandsma RHJ, Koulman A, Thompson DS. Weight gain during nutritional rehabilitation post-childhood malnutrition may influence the associations between adulthood desaturases activity and anthro-cardiometabolic risk factors. Clin Nutr 2024; 43:747-755. [PMID: 38330703 DOI: 10.1016/j.clnu.2024.01.032] [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: 08/24/2023] [Revised: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUNDS & AIMS Childhood malnutrition is a major global health problem with long-term sequelae, including non-communicable diseases (NCDs). Mechanisms are unknown but may involve metabolic programming, resulting from "short-term" solutions to optimise survival by compromising non-priority organs. As key players in lipid metabolism, desaturases have been shown to be predictive of NCDs. We hypothesised that the association between specific desaturase activities and NCD risk determinants (including body composition, serum glucose, insulin levels, and blood pressure) are influenced by childhood post-malnutrition weight gain. METHODS 278 Afro-Caribbean adults with well-documented clinical history of severe malnutrition in childhood were studied. Extensive metabolic analyses including body composition (DXA), fasting serum glucose and lipidomics (n = 101), and fasting serum insulin (n = 83) were performed in malnutrition survivors and matched community controls (n = 90). Established lipid ratios were used as proxies of desaturase activities: CE 16:1/CE 16:0 for stearoyl-CoA desaturase (SCD1), LysoPC 20:4/20:3 for fatty acid desaturase 1 (FADS1), and LysoPC 20:3/18:2 for FADS2. RESULTS Compared to community controls, adult malnutrition survivors (mean ± SD) age 28.3 ± 7.8 and BMI 23.6 ± 5.2 had higher SCD1 and FADS1 activity, (B ± SE) 0.07 ± 0.02 and 0.7 ± 0.08, respectively, but lower FADS2 activities (B ± SE) -0.05 ± 0.01, adjusted for sex and age (p < 0.0005). SCD1 was positively associated with adult BMI and body fat percentage, and negatively associated with lean mass and height. Stratification based on weight gain during nutritional rehabilitation among malnutrition survivors might signal the potential associations between weight gain during that critical period, desaturase activities, and some of adult metabolic parameters, with the lowest tertiles (slowest catch-up weight gain) performing more similarly to controls. CONCLUSIONS In adult survivors of early-life severe acute malnutrition, desaturase activity is associated with markers of NCD risk, especially adiposity. These associations seem to be strengthened by faster weight gain during nutritional rehabilitation.
Collapse
Affiliation(s)
- Laurentya Olga
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
| | - Kimberley McKenzie
- Caribbean Institute for Health Research, The University of the West Indies, Kingston, Jamaica
| | - Marko Kerac
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Michael Boyne
- Department of Medicine, The University of the West Indies, Kingston, Jamaica
| | - Asha Badaloo
- Caribbean Institute for Health Research, The University of the West Indies, Kingston, Jamaica
| | - Robert H J Bandsma
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada; Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Albert Koulman
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK; Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Debbie S Thompson
- Caribbean Institute for Health Research, The University of the West Indies, Kingston, Jamaica
| |
Collapse
|
26
|
Dehghan Harati HR, Mobedi E, Allahyari I, Gharagozlou F, Vojgani M, Hemmati Baghbanani R, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: III. Association of level of maternal milk production with offspring's birth weight, survival, productive and reproductive performance and AMH concentration from birth to the first lactation period. Theriogenology 2024; 216:155-167. [PMID: 38183932 DOI: 10.1016/j.theriogenology.2024.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Although some studies investigated the relationship of dam milk production (DMP) with offspring birth weight and productive performance, limited information is available on the association of level of DMP with reproductive performance in dairy cows. Therefore, the present study was conducted to understand whether dams with various levels of milk production produce offspring with different fertility. In study I, offspring were classified based on the level of DMP into five categories, including DMP1 (dams with <10.00 × 103 kg of 305-day milk production), DMP2 (dams with ≥10.00 × 103 kg and <12.00 × 103 kg of 305-day milk production), DMP3 (dams with ≥12.00 × 103 kg and <14.00 × 103 kg of 305-day milk production), DMP4 (dams with ≥14.00 × 103 kg and <16.00 × 103 kg of 305-day milk production) and DMP5 (dams with ≥16.00 × 103 kg of 305-day milk production). In study I, data of birth weight, milk yield and reproductive variables of 14,536 offspring born to lactating dams and corresponding data of DMP were retrieved. In study II, blood samples (n = 339) were collected from offspring in various categories of DMP for measurement of serum AMH. Offspring were heavier at birth in DMP4 and DMP5 categories than DMP1 and DMP2 categories (P < 0.05). Milk yield of offspring increased as DMP elevated (P < 0.05); however, offspring in DMP1 and DMP2 categories produced higher milk as compared with their dams during primiparity (P < 0.05) whereas offspring in DMP3, DMP4 and DMP5 categories produced less milk as compared with their dams during primiparity (P < 0.05). Milk fat to protein ratio during the first month of lactation was greater in DMP4 and DMP5 categories than DMP1 category (P < 0.05). Offspring of DMP4 and DMP5 categories were inseminated and conceived at younger ages than offspring of DMP1 category during nulliparity (P < 0.05). Calving to conception interval was longer in DMP5 than DMP1 category in primiparous offspring (P < 0.05), but concentration of AMH did not differ among various categories of DMP (P > 0.05). In conclusion, dams with greater level of milk production produced heavier offspring with higher milk yield but worse transgenerational improvement in milk production and diminished reproductive performance, which were seemingly under higher pressure of negative energy balance during the first month of lactation.
Collapse
Affiliation(s)
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
27
|
Tain YL, Hsu CN. Nutritional Approaches Targeting Gut Microbiota in Oxidative-Stress-Associated Metabolic Syndrome: Focus on Early Life Programming. Nutrients 2024; 16:683. [PMID: 38474810 DOI: 10.3390/nu16050683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolic syndrome (MetS) denotes a constellation of risk factors associated with the development of cardiovascular disease, with its roots potentially traced back to early life. Given the pivotal role of oxidative stress and dysbiotic gut microbiota in MetS pathogenesis, comprehending their influence on MetS programming is crucial. Targeting these mechanisms during the early stages of life presents a promising avenue for preventing MetS later in life. This article begins by examining detrimental insults during early life that impact fetal programming, ultimately contributing to MetS in adulthood. Following that, we explore the role of oxidative stress and the dysregulation of gut microbiota in the initiation of MetS programming. The review also consolidates existing evidence on how gut-microbiota-targeted interventions can thwart oxidative-stress-associated MetS programming, encompassing approaches such as probiotics, prebiotics, postbiotics, and the modulation of bacterial metabolites. While animal studies demonstrate the favorable effects of gut-microbiota-targeted therapy in mitigating MetS programming, further clinical investigations are imperative to enhance our understanding of manipulating gut microbiota and oxidative stress for the prevention of MetS.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
28
|
Her TK, Li J, Lin H, Liu D, Root KM, Regal JF, Alejandro EU, Cao R. Circadian Disruption across Lifespan Impairs Glucose Homeostasis and Insulin Sensitivity in Adult Mice. Metabolites 2024; 14:126. [PMID: 38393018 PMCID: PMC10892663 DOI: 10.3390/metabo14020126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Circadian rhythm disruption is associated with impaired glucose homeostasis and type 2 diabetes. For example, night shift work is associated with an increased risk of gestational diabetes. However, the effects of chronic circadian disruption since early life on adult metabolic health trajectory remain unknown. Here, using the "Short Day" (SD) mouse model, in which an 8 h/8 h light/dark (LD) cycle was used to disrupt mouse circadian rhythms across the lifespan, we investigated glucose homeostasis in adult mice. Adult SD mice were fully entrained into the 8 h/8 h LD cycle, and control mice were entrained into the 12 h/12 h LD cycle. Under a normal chow diet, female and male SD mice displayed a normal body weight trajectory. However, female but not male SD mice under a normal chow diet displayed glucose intolerance and insulin resistance, which are associated with impaired insulin signaling/AKT in the skeletal muscle and liver. Under high-fat diet (HFD) challenges, male but not female SD mice demonstrated increased body weight gain compared to controls. Both male and female SD mice developed glucose intolerance under HFD. Taken together, these results demonstrate that environmental disruption of circadian rhythms contributes to obesity in a sexually dimorphic manner but increases the risk of glucose intolerance and insulin resistance in both males and females.
Collapse
Affiliation(s)
- Tracy K. Her
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Jin Li
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Hao Lin
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Dong Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Kate M. Root
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
| | - Jean F. Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
| | - Emilyn U. Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (J.L.); (H.L.); (D.L.); (K.M.R.); (J.F.R.)
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
29
|
Wells JCK, Desoye G, Leon DA. Reconsidering the developmental origins of adult disease paradigm: The 'metabolic coordination of childbirth' hypothesis. Evol Med Public Health 2024; 12:50-66. [PMID: 38380130 PMCID: PMC10878253 DOI: 10.1093/emph/eoae002] [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: 08/22/2023] [Revised: 12/18/2023] [Indexed: 02/22/2024] Open
Abstract
In uncomplicated pregnancies, birthweight is inversely associated with adult non-communicable disease (NCD) risk. One proposed mechanism is maternal malnutrition during pregnancy. Another explanation is that shared genes link birthweight with NCDs. Both hypotheses are supported, but evolutionary perspectives address only the environmental pathway. We propose that genetic and environmental associations of birthweight with NCD risk reflect coordinated regulatory systems between mother and foetus, that evolved to reduce risks of obstructed labour. First, the foetus must tailor its growth to maternal metabolic signals, as it cannot predict the size of the birth canal from its own genome. Second, we predict that maternal alleles that promote placental nutrient supply have been selected to constrain foetal growth and gestation length when fetally expressed. Conversely, maternal alleles that increase birth canal size have been selected to promote foetal growth and gestation when fetally expressed. Evidence supports these hypotheses. These regulatory mechanisms may have undergone powerful selection as hominin neonates evolved larger size and encephalisation, since every mother is at risk of gestating a baby excessively for her pelvis. Our perspective can explain the inverse association of birthweight with NCD risk across most of the birthweight range: any constraint of birthweight, through plastic or genetic mechanisms, may reduce the capacity for homeostasis and increase NCD susceptibility. However, maternal obesity and diabetes can overwhelm this coordination system, challenging vaginal delivery while increasing offspring NCD risk. We argue that selection on viable vaginal delivery played an over-arching role in shaping the association of birthweight with NCD risk.
Collapse
Affiliation(s)
- Jonathan C K Wells
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
| | - David A Leon
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
30
|
Rimbach R, Pontzer H. Increased physical activity is not related to markers of cardiometabolic health in two lemur species. Am J Primatol 2024; 86:e23564. [PMID: 37839049 DOI: 10.1002/ajp.23564] [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: 08/08/2023] [Revised: 09/18/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Abstract
Insufficient physical activity is a major risk factor for cardiometabolic disease (i.e., unhealthy weight gain, heart disease, and diabetes) in humans and may also negatively affect health of primates in human care. Effects of physical activity on energy expenditure and cardiometabolic health are virtually unstudied in nonhuman primates. We investigated physical activity and metabolic markers in 15 adult ring-tailed lemurs (Lemur catta) and 11 Coquerel's sifakas (Propithecus coquereli) at the Duke Lemur Center during a period of low activity in winter when the animals were housed in buildings (with outdoor access) and a period of high activity when individuals were free-ranging in large, outdoor, forested enclosures. We compared body mass, blood glucose, triglycerides, HDL- and LDL-cholesterol, physical activity via accelerometry, and total energy expenditure (TEE) via the doubly labeled water method (in ring-tailed lemurs only) between both conditions. Both species were more active and had a lower body mass in summer. Ring-tailed lemurs had a higher TEE and lower triglyceride levels in summer, whereas sifaka had higher triglyceride levels in summer. Individuals that increased their activity more, also lost more body mass. Individuals that lost more body mass, also had a positive change in HDL-cholesterol (i.e., higher values in summer). Changes in activity were not associated with changes in markers of metabolic health, body fat percentage and TEE (both unadjusted and adjusted for body composition). Older age was associated with lower activity in both species, and decreased glucose in ring-tailed lemurs, but was otherwise unrelated to metabolic markers and, for ring-tailed lemurs, adjusted TEE. Overall, body mass was lower during summer but the increase in physical activity did not strongly influence metabolic health or TEE in these populations.
Collapse
Affiliation(s)
- Rebecca Rimbach
- Evolutionary Anthropology, Duke University, Durham, North Carolina, USA
- Department of Behavioural Biology, University of Münster, Münster, Germany
- School of Animal, Plant & Environmental Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Herman Pontzer
- Evolutionary Anthropology, Duke University, Durham, North Carolina, USA
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
31
|
Saeed H, Wu J, Tesfaye M, Grantz KL, Tekola-Ayele F. Placental accelerated aging in antenatal depression. Am J Obstet Gynecol MFM 2024; 6:101237. [PMID: 38012987 PMCID: PMC10843762 DOI: 10.1016/j.ajogmf.2023.101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Antenatal maternal depression is associated with poor pregnancy outcomes and long-term effects on the offspring. Previous studies have identified links between antenatal depression and placental DNA methylation and between placental epigenetic aging and poor pregnancy outcomes, such as preterm labor and preeclampsia. The relationship between antenatal depression and poor pregnancy outcomes may be partly mediated via placental aging. OBJECTIVE This study aimed to investigate whether antenatal depressive symptoms are associated with placental epigenetic age acceleration, an epigenetic aging clock measure derived from the difference between methylation age and gestational age at delivery. STUDY DESIGN The study included 301 women who provided placenta samples at delivery as part of the Eunice Kennedy Shriver National Institute of Child Health and Human Development Fetal Growth Studies - Singletons that recruited participants from diverse race and ethnic groups at 12 US clinical sites (2009-2013). Women underwent depression screening using the Edinburgh Postnatal Depression Scale up to 6 times across the 3 trimesters of pregnancy. Depressive symptoms status was determined for each pregnancy trimester using an Edinburgh Postnatal Depression Scale score, in which a score of ≥10 was defined as having depressive symptoms and a score of <10 was defined as not having depressive symptoms. Placental DNA methylation was profiled from placenta samples. Placental epigenetic age was estimated using a methylation-based age estimator (placental "epigenetic clock") that has previously been found to have high placental gestational age prediction accuracy for uncomplicated term pregnancies. Placental age acceleration was defined to be the residual upon regressing the estimated epigenetic age on gestational age at delivery. Associations between an Edinburgh Postnatal Depression Scale score of ≥10 and an Edinburgh Postnatal Depression Scale score of <10 in the first, second, and third trimesters of pregnancy (ie, depressive symptoms vs none in each trimester) and placental age acceleration were tested using multivariable linear regression adjusting for maternal age, parity, race and ethnicity, and employment. RESULTS There were 31 (10.3%), 48 (16%), and 49 (16.4%) women with depressive symptoms (ie, Edinburgh Postnatal Depression Scale score of ≥10) in the first, second, and third trimesters of pregnancy, respectively. Of these women, 21 (7.2%) had sustained first- and second-trimester depressive symptoms, 19 (7%) had sustained second- and third-trimester depressive symptoms, and 12 (4.8%) had sustained depressive symptoms throughout pregnancy. Women with depressive symptoms in the second trimester of pregnancy had 0.41 weeks higher placental age acceleration than women without depressive symptoms during the second trimester of pregnancy (β=0.21 weeks [95% confidence interval, -0.17 to 0.58; P=.28] during the first trimester of pregnancy; β=0.41 weeks [95% confidence interval, 0.10-0.71; P=.009] during the second trimester of pregnancy; β=0.17 weeks [95% confidence interval, -0.14 to 0.47; P=.29] during the third trimester of pregnancy). Sustained first- and second-trimester depressive symptoms were associated with 0.72 weeks higher placental age acceleration (95% confidence interval, 0.29-1.15; P=.001) than no depressive symptom in the 2 trimesters. The association between second-trimester depressive symptoms and higher placental epigenetic age acceleration strengthened in the analysis of pregnancies with male fetuses (β=0.53 weeks; 95% confidence interval, 0.06-1.08; P=.03) but was not significant in pregnancies with female fetuses. CONCLUSION Antenatal depressive symptoms during the second trimester of pregnancy were associated with an average of 0.41 weeks of increased placental age acceleration. Accelerated placental aging may play an important role in the underlying mechanism linking antenatal depression to pregnancy complications related to placental dysfunction.
Collapse
Affiliation(s)
- Haleema Saeed
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD (Dr Saeed); Department of Maternal-Fetal Medicine, Medstar Washington Hospital Center, Washington, DC (Drs Saeed)
| | - Jing Wu
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Markos Tesfaye
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, University of Oslo, Oslo, Norway (Dr Tesfaye); Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Dr Tesfaye)
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele).
| |
Collapse
|
32
|
Davidson B, Gonzales E, Mast G, Laporta J. Late-gestation heat stress in Holstein dams programs in utero development of daughter's germline, triggering skin and hair morphology adaptations of granddaughters. JDS COMMUNICATIONS 2024; 5:83-88. [PMID: 38223391 PMCID: PMC10785235 DOI: 10.3168/jdsc.2023-0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/09/2023] [Indexed: 01/16/2024]
Abstract
Homeostasis and thermoregulation are influenced by the interplay of hair coat and skin characteristics. Our previous work indicated that hair and skin adaptations, triggered by in utero heat stress, affect thermoregulation in postnatal life. Herein, we investigate multigenerational carry-over effects of late-gestation heat stress on hair and skin characteristics beyond the first generation. Pregnant Holstein dams (F0, grand-dams) were heat stressed (HT, shade, n = 41) or provided active cooling (CL, shade, fans, and water soakers, n = 41) for the last 56 d of gestation during summer (temperature-humidity index ≥68). The first generation of heifers (F1, daughters) who were in utero heat stressed (HTF1, n = 36) or not (CLF1, n = 37) were born and raised as a cohort from birth to first calving. Thirty second-generation heifers (F2, granddaughters), born to HTF1 (HTF2, n = 12) and CLF1 (CLF2, n = 18), were raised as a cohort from birth to 70 d of age. Hair samples and skin biopsies from HTF2 and CLF2 were collected on postnatal d 70 (n = 6/group). Hair samples were subdivided into topcoat and undercoat, and skin tissue was fixed for ~18 h in 10% formalin, dehydrated, paraffin-embedded, sectioned, and stained with hematoxylin and eosin to visualize morphology. Variables analyzed included hair length and diameter; stratum corneum cross-sectional area and thickness; epidermis thickness; sweat gland depth, number, cross-sectional area, and average size; and sebaceous gland number, cross-sectional area, and average size. Measurements were performed using the ImageJ software and analyzed using PROC MIXED in SAS (SAS Institute Inc.). Hair length and diameter tended to be shorter and thicker in HTF2, compared with CLF2. The HTF2 skin had smaller stratum corneum cross-sectional area and tended to a thinner epidermis. to CLF2, HTF2 skin had more but smaller sebaceous glands, whereas no differences in sweat glands were observed. In summary, we report phenotypic alterations in hair and skin characteristics of granddaughters. Whether these adaptations grant improved postnatal thermoregulatory ability for the granddaughters remains to be investigated.
Collapse
Affiliation(s)
- B.D. Davidson
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706
| | - E.T. Gonzales
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706
| | - G.L. Mast
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706
| | - J. Laporta
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706
| |
Collapse
|
33
|
Lee E, Lee SY, Kim HB, Yang SI, Yoon J, Suh DI, Oh HY, Ahn K, Kim KW, Shin YH, Hong SJ. Insights from the COCOA birth cohort: The origins of childhood allergic diseases and future perspectives. Allergol Int 2024; 73:3-12. [PMID: 37752021 DOI: 10.1016/j.alit.2023.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
The ongoing COhort for Childhood Origin of Asthma and allergic diseases (COCOA) study is a prospective birth cohort investigating the origin and natural courses of childhood allergic diseases, including atopic dermatitis, food allergy, allergic rhinitis and asthma, with long-term prognosis. Initiated under the premise that allergic diseases result from a complex interplay of immune development alterations, environmental exposures, and host susceptibility, the COCOA study explores these dynamic interactions during prenatal and postnatal periods, framed within the hygiene and microbial hypotheses alongside the developmental origins of health and disease (DOHaD) hypothesis. The scope of the COCOA study extends to genetic predispositions, indoor and outdoor environmental variables affecting mothers and their offsprings such as outdoor and indoor air pollution, psychological factors, diets, and the microbiomes of skin, gut, and airway. We have embarked on in-depth investigations of diverse risk factors and the pathophysiological underpinnings of allergic diseases. By employing multi-omics approaches-proteomics, transcriptomics, and metabolomics-we gain deeper insights into the distinct pathophysiological processes across various endotypes of childhood allergic diseases, incorporating the exposome using extensive resources within the COCOA study. Integration with large-scale datasets, such as national health insurance records, enhances robustness and mitigates potential limitations inherent to birth cohort studies. As part of global networks focused on childhood allergic diseases, the COCOA study fosters collaborative research across multiple cohorts. The findings from the COCOA study are instrumental in informing precision medicine strategies for childhood allergic diseases, underpinning the establishment of disease trajectories.
Collapse
Affiliation(s)
- Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, South Korea
| | - So-Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo-Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, South Korea
| | - Song-I Yang
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Jisun Yoon
- Department of Pediatrics, Chung-Ang University College of Medicine, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea
| | - Dong In Suh
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Hea Young Oh
- Department of Medicine, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Youn Ho Shin
- Department of Pediatrics, The Catholic University of Korea, Yeouido St. Mary's Hospital, Seoul, South Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
34
|
Jing Jia, Ma B, Zhao X. Fetal endothelial colony-forming cells: Possible targets for prevention of the fetal origins of adult diseases. Placenta 2024; 145:80-88. [PMID: 38100962 DOI: 10.1016/j.placenta.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Endothelial colony-forming cells (ECFCs), a subset of circulating and resident endothelial progenitor cells, are capable of self-renewal and de novo vessel formation, and are known key regulators of vascular integrity and homeostasis. Numerous studies have found that exposure to hostile environment during the fetal development exerts a profound influence on the level and function of ECFCs, which may be the underlying factor linking endothelial dysfunction to cardiovascular disease of the offspring in later life. Herein, we focus on the latest findings regarding the effects of pregnancy-related disorders on the frequency and function of fetal ECFCs. Subsequently, we discuss about placental ECFCs and put forward some details that should be paid attention to in the process of ECFC isolation and culture. Overall, the information presented in this review highlight the potential of ECFCs as a future biomarker or even therapeutic targets for the pregnancy-related adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Jing Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Baitao Ma
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
35
|
Mozaffari Makiabadi MJ, Bafandeh M, Gharagozlou F, Vojgani M, Mobedi E, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: II. Association of gestational stage of maternal exposure to heat stress with offspring's birth weight, milk yield, reproductive performance and AMH concentration during the first lactation period. Theriogenology 2023; 212:41-49. [PMID: 37690376 DOI: 10.1016/j.theriogenology.2023.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Although the negative effect of maternal exposure to heat stress on production and reproduction of offspring has been reported, there are some discrepancies among various studies about which gestational stage is more critical in this regard. Therefore, the present research was conducted to identify during which stage(s) of pregnancy maternal exposure to heat stress would lead to more dramatic decrease in productive and reproductive performance of offspring. To this end, offspring were classified based on the gestational stage they were in utero exposed to heat stress into four categories, including heat stress exposure (HSE) during only the first trimester of gestation (HSE1), HSE during the first and second trimester of gestation (HSE2), HSE during the second and third trimester of gestation (HSE3) and HSE during only the third trimester of gestation (HSE4). In study I, data of birth weight, milk yield and reproductive variables of 11,788 offspring and data of the month they were conceived were retrieved. In study II, blood samples (n = 521) were collected from offspring in various categories of HSE for measurement of serum AMH. Offspring in HSE1 and HSE2 categories were heavier than offspring in HSE3 and HSE4 categories (P < 0.0001). Offspring in HSE1 and HSE3 categories had the lowest and highest milk production, respectively (P < 0.05). First service conception rate was the greatest and worst in HSE1 and HSE4 categories, respectively (P < 0.05). Service per conception and calving to conception interval were greater in HSE2 than HSE4 category (P < 0.05). Concentration of AMH was lower in HSE1 than HSE4 category (P < 0.05). In conclusion, the present study indicated that the early stage of gestation could be a more critical period for the negative impact of in utero heat stress on developmental programming of milk production and ovarian reserves. Yet an evident temporal pattern for the adverse effect of maternal heat stress on developmental programming of reproductive performance in offspring was not found.
Collapse
Affiliation(s)
| | - Mohammad Bafandeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
36
|
Gleason JL, Yeung EH, Sundaram R, Putnick DL, Mendola P, Bell EM, Polinski KJ, Robinson SL, Grantz KL. Longitudinal Child Growth Patterns in Twins and Singletons in the Upstate KIDS Cohort. J Pediatr 2023; 263:113720. [PMID: 37660974 PMCID: PMC10872829 DOI: 10.1016/j.jpeds.2023.113720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVES To investigate childhood growth patterns in twins and to determine whether they show the same signs of excess growth as singletons born small-for-gestational age (SGA), which may confer future cardiometabolic risk. STUDY DESIGN In the Upstate KIDS cohort of infants delivered from 2008 through 2010, we compared height, weight, and body mass index (BMI) z-scores at 0-3 and 7-9 years of age, as well as risk of rapid weight gain (RWG) in infancy and overweight/obesity beginning at 2 years, among appropriate-for-gestational age (AGA) twins (n = 1121), AGA singletons (n = 2684), and two groups of SGA twins: uncertain SGA twins (<10th percentile for birthweight by a singleton reference but >10th% by a population-based twin birthweight reference; n = 319) and true SGA twins (<10th% by a population-based twin reference; n = 144). RESULTS Compared with AGA twins, both SGA twin groups had lower weight and BMI z-scores at both time points. By 7-9 years, both groups caught up in height with AGA twins. Compared with AGA singletons, z-score differences decreased between 0-3 and 7-9 years for uncertain SGA and true SGA twins, though true SGA twins had the lowest z-scores for all measures. During infancy, twins were more likely to display RWG compared with AGA singletons (RR = 2.06 to 2.67), which may reflect normal catch-up growth, as no twin group had higher prevalence of overweight/obesity at either time point. CONCLUSIONS Though twins had lower height, weight, and BMI z-scores at birth and into toddlerhood, differences were reduced by 7-9 years, with no evidence of pathological growth and no group of twins showing elevated risk of overweight/obesity.
Collapse
Affiliation(s)
- Jessica L Gleason
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Edwina H Yeung
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Rajeshwari Sundaram
- Biostatistics and Bioinformatics Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Diane L Putnick
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Pauline Mendola
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY
| | - Erin M Bell
- Departments of Environmental Health Sciences and Epidemiology and Biostatistics University at Albany, School of Public Health, Rensselaer, NY
| | - Kristen J Polinski
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Sonia L Robinson
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD.
| |
Collapse
|
37
|
Pieszka M, Szczurek P, Orczewska-Dudek S, Kamyczek M, Pieszka M. Determining the Effect of Pancreatic-like Enzymes (PLEMs) Added to the Feed of Pregnant Sows on Fetal Size of Piglets to Minimize IUGR Syndrome Caused by Fetal Malnutrition. Animals (Basel) 2023; 13:3448. [PMID: 38003066 PMCID: PMC10668663 DOI: 10.3390/ani13223448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
The present study aimed to develop a feeding strategy for pregnant sows that involved the prenatal administration of a mixture of pancreatic-like fungal enzymes, such as lipase, amylase, and protease, at (1) 1-115 days of gestation (group D1) and (2) 80-115 days of gestation (group D2) and to carry out a comparison with groups of sows that were not receiving such supplementation (negative control (NC) and positive control (PC)). It was found that the administration of the enzyme supplement resulted in a significant shortening of gestation (p ≤ 0.01). The pancreatic enzymes administered to sows had a significant effect on the number of liveborn piglets and weaned piglets, which was higher compared with the control groups that did not receive supplementation: D1-12.1 ± 1.1 and 11.12 ± 1.1 and D2-12.8 ± 1.3 and 11.75 ± 0.07 vs. the control groups KN-10.7 ± 1.0 and 9.62 ± 0.95 and KP-10.9 ± 1.2 and 10.15 ± 1.0 (p < 0.006), respectively. Significant changes in piglet growth were observed after weaning up to 70 days of age. During this period, the most favorable growth parameters were observed in groups D2 (420 ± 91 g) and PC (407 ± 103 g), in which piglets obtained a mixture of pancreatic enzymes (lipase, amylase, and protease) at 3 weeks of age, and significantly higher weight gain and feed intake were observed compared with groups NC (378 ± 114 g) and D1 (381 ± 96 g) (p ≤ 0.007). In contrast, insulin levels were significantly lower in groups D1 and D2, with values of 6.8 IU/mL and 6.7 IU/mL, respectively, compared with groups NC (14.6 IU/mL) and PC (16.6 IU/mL) (p ≤ 0.01). Piglets in group D2 had a significantly better feed conversion ratio (FCR) of 1.604 ± 0.10 compared with the other dietary groups: KN-1.986 ± 0.14; KP-1.704 ± 0.11; and D1-1.932 ± 0.15 (p ≤ 0.03). Histological imaging confirmed a significantly thicker intestinal epithelium and intestinal mesenteron in animals from groups D2 and PC (p ≤ 0.03). Animals from the groups KP, D1, and D2 receiving enzymes showed a highly significant increase in the surface area of pancreatic follicles and pancreatic surface area compared with the group without KN supplementation (p < 0.01). Furthermore, significantly higher activity of the brush border enzyme lactase was observed in groups D1, D2, and PC, with values of 32.90 ± 3.99, 30.00 ± 6.83, and 29.60 ± 29.60, respectively, compared with group NC, with a value of 21.80 ± 3.27 (p ≤ 0.01).
Collapse
Affiliation(s)
- Marek Pieszka
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, 32-083 Kraków, Poland (S.O.-D.)
| | - Paulina Szczurek
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, 32-083 Kraków, Poland (S.O.-D.)
| | - Sylwia Orczewska-Dudek
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, 32-083 Kraków, Poland (S.O.-D.)
| | - Marian Kamyczek
- National Research Institute of Animal Production, The Institute’s Experimental Station, Mielżynskich Street 14, 64-122 Pawłowice, Poland;
| | - Magdalena Pieszka
- Department of Animal Genetics, Breeding and Ethology, University of Agriculture, Mickiewicza 24/28, 30-059 Kraków, Poland;
| |
Collapse
|
38
|
Gou W, Wang H, Tang XY, He Y, Su C, Zhang J, Sun TY, Jiang Z, Miao Z, Fu Y, Zhao H, Chen YM, Zhang B, Zhou H, Zheng JS. Early-life exposure to the Great Chinese Famine and gut microbiome disruption across adulthood for type 2 diabetes: three population-based cohort studies. BMC Med 2023; 21:414. [PMID: 37907866 PMCID: PMC10619253 DOI: 10.1186/s12916-023-03123-y] [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: 06/07/2023] [Accepted: 10/20/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND The early life stage is critical for the gut microbiota establishment and development. We aimed to investigate the lifelong impact of famine exposure during early life on the adult gut microbial ecosystem and examine the association of famine-induced disturbance in gut microbiota with type 2 diabetes. METHODS We profiled the gut microbial composition among 11,513 adults (18-97 years) from three independent cohorts and examined the association of famine exposure during early life with alterations of adult gut microbial diversity and composition. We performed co-abundance network analyses to identify keystone taxa in the three cohorts and constructed an index with the shared keystone taxa across the three cohorts. Among each cohort, we used linear regression to examine the association of famine exposure during early life with the keystone taxa index and assessed the correlation between the keystone taxa index and type 2 diabetes using logistic regression adjusted for potential confounders. We combined the effect estimates from the three cohorts using random-effects meta-analysis. RESULTS Compared with the no-exposed control group (born during 1962-1964), participants who were exposed to the famine during the first 1000 days of life (born in 1959) had consistently lower gut microbial alpha diversity and alterations in the gut microbial community during adulthood across the three cohorts. Compared with the no-exposed control group, participants who were exposed to famine during the first 1000 days of life were associated with consistently lower levels of keystone taxa index in the three cohorts (pooled beta - 0.29, 95% CI - 0.43, - 0.15). Per 1-standard deviation increment in the keystone taxa index was associated with a 13% lower risk of type 2 diabetes (pooled odds ratio 0.87, 95% CI 0.80, 0.93), with consistent results across three individual cohorts. CONCLUSIONS These findings reveal a potential role of the gut microbiota in the developmental origins of health and disease (DOHaD) hypothesis, deepening our understanding about the etiology of type 2 diabetes.
Collapse
Affiliation(s)
- Wanglong Gou
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Huijun Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, China
| | - Xin-Yi Tang
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yan He
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chang Su
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, China
| | - Jiguo Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, China
| | - Ting-Yu Sun
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Zengliang Jiang
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Zelei Miao
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yuanqing Fu
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Hui Zhao
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China.
| | - Bing Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China.
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, China.
| | - Hongwei Zhou
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Ju-Sheng Zheng
- Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- School of Life Sciences, Westlake University, Hangzhou, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
39
|
Quirici V, Valeris-Chacín CE, Parada P, Cuevas E, Wingfield JC. Baseline Corticosterone, Stress Responses, and Leukocyte Profiles in Chicks of Precocial Birds in Rural and Urban Environments. Life (Basel) 2023; 13:2138. [PMID: 38004278 PMCID: PMC10672541 DOI: 10.3390/life13112138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
The urban environment produces complex relationship among urban stressors that could change the levels of the steroid hormone, glucocorticoid (GCs). Studies that have evaluated baseline corticosterone (Cort) levels (the main GC in birds) and stress responses during development in urban and rural environments have obtained contrasting results. This ambiguity could partially be because the studies were carried out in altricial species, where parental care and sibling competition can affect Cort levels. Therefore, in this study, we compared levels of circulating baseline levels of CORT (blood sample obtained within 3 min of capture) and stress responses (blood sample obtained 30 min after capture) and the H/L ratio (an alternative method to measure stress) in chicks of a precocial bird, southern lapwings (Vanellus chilensis), from one rural (6 chicks), one urban low-polluted (13 chicks), and one urban high-polluted (10 chicks) site of Metropolitan Region of Santiago de Chile. We observed higher baseline Cort (2.41 ± 1.78 ng/mL) in the urban high-polluted site, a higher H/L ratio (0.51 ± 0.20) in the urban low-polluted site, and similar stress response across the three sites. We propose that the difference in stress physiology we observed within Santiago de Chile is because the two zones are at extremes in terms of stressors (noise, light, chemical, and human presence). It is unusual to find a precocious bird that lives in both urban and rural areas; therefore, the results of this study will advance our knowledge of the effect of the urban environment during the development of wildlife, which is relevant in terms of management and conservation.
Collapse
Affiliation(s)
- Verónica Quirici
- Centro de Investigación para la Sustentabilidad, Facultad de Ciencias de la Vida, Universidad Andres Bello, Av. República 440, Santiago 8370251, Chile
| | - Carlos E. Valeris-Chacín
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago 8330015, Chile
- Cape Horn International Center for Global Change Studies and Biocultural Conservation (CHIC), O’Higgins 310, Cabo de Hornos, Puerto Williams 6350000, Chile
| | - Pablo Parada
- Centro de Investigación para la Sustentabilidad, Facultad de Ciencias de la Vida, Universidad Andres Bello, Av. República 440, Santiago 8370251, Chile
| | - Elfego Cuevas
- Centro de Investigación para la Sustentabilidad, Facultad de Ciencias de la Vida, Universidad Andres Bello, Av. República 440, Santiago 8370251, Chile
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Quillota 980, Viña del Mar 2520000, Chile
| | - John C. Wingfield
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
40
|
Bafandeh M, Mozaffari Makiabadi MJ, Gharagozlou F, Vojgani M, Mobedi E, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: I. Association of maternal parity with offspring's birth weight, milk yield, reproductive performance and AMH concentration during the first lactation period. Theriogenology 2023; 210:34-41. [PMID: 37473594 DOI: 10.1016/j.theriogenology.2023.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
Multiparous dams have been reported to produce offspring with greater fertility and higher AMH concentration, as a marker of ovarian reserves, as compared with nulliparous and primiparous dams. Yet it has remained to be addressed whether this phenomenon can still be true for old multiparous cows which might experience some geriatric changes in their reproductive system. Therefore, the present study was conducted to evaluate the productive and reproductive performance of offspring with different maternal parity. To this end, offspring were classified based on their maternal parities into four categories, including offspring of nulliparous (no previous parity), primiparous (one previous parity), young multiparous (two to six previous parities) and old multiparous (seven or more previous parities) dams. In study I, data of birth weight, milk yield and reproductive variables of 11,788 offspring and data of their maternal parity were retrieved. In study II, blood samples (n = 521) were collected from offspring with various maternal parity for measurement of serum AMH. Birth weight was the lowest in the offspring of nulliparous dams (P < 0.0001) and it was lower in offspring of primiparous and old multiparous dams than offspring of young multiparous dams (P < 0.05). Milk production was the lowest in offspring of old multiparous dams (P < 0.01), and it was lower in offspring of young multiparous dams than offspring of nulliparous and primiparous dams (P < 0.0001). Offspring of old multiparous dams had greater first service conception rate, less services per conception and shorter calving to conception interval than offspring of nulliparous, primiparous and young multiparous dams (P < 0.05). Furthermore, AMH concentration was higher in offspring of old multiparous dams than offspring of nulliparous and primiparous dams (P < 0.05). In conclusion, the present study revealed greater milk production in offspring resulting from dams with lower parity, probably due to the genetic selection for improvement of milk production in dairy cows which imparts the younger generations greater genetic merits for milk production. Reproductive performance, however, was greater in offspring born to dams with higher parity, particularly those born to old multiparous dams, and this phenomenon might be related to their lower milk production and higher AMH concentration.
Collapse
Affiliation(s)
- Mohammad Bafandeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
41
|
Yonke JA, Seymour KA, El-Kadi SW. Branched-chain amino acid supplementation does not enhance lean tissue accretion in low birth weight neonatal pigs, despite lower Sestrin2 expression in skeletal muscle. Amino Acids 2023; 55:1389-1404. [PMID: 37743429 DOI: 10.1007/s00726-023-03319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023]
Abstract
Postnatal muscle growth is impaired in low birth weight (L) neonatal pigs. Leucine supplementation has been established as a dietary intervention to enhance muscle growth in growing animals. The aim of this study was to investigate the efficacy of supplementing L neonatal pig formulas with branched-chain amino acids (B) to enhance the rate of protein accretion. Twenty-four 3-day old pigs were divided into two groups low (L) and normal birth weight (N) based on weight at birth. Pigs were assigned to a control (C) or 1% branched-chain amino acids (B) formulas, and fed at 250 mL·kg body weight -1·d-1 for 28 d. Body weight of pigs in the L group was less than those in the N group (P < 0.01). However, fractional body weight was greater for L pigs compared with their N siblings from day 24 to 28 of feeding regardless of formula (P < 0.01). In addition, feed efficiency (P < 0.0001) and efficiently of protein accretion (P < 0.0001) were greater for L than N pigs regardless of supplementation. Pigs fed the B formula had greater plasma leucine, isoleucine, and valine concentrations compared with those fed the C formula (P < 0.05). Longissimus dorsi Sestrin2 protein expression was less for pigs in the L group compared with those in the N group (P < 0.01), but did not result in a corresponding increase in translation initiation signaling. Longissimus dorsi mRNA expression of BCAT2 was less for LB pigs compared with those in the LC group, and was intermediate for NC and NB pigs (P < 0.05). Hepatic mRNA expression of BCKDHA was greater for pigs in the L compared with those in the N groups (P < 0.05). However, plasma branched-chain keto-acid concentration was reduced for C compared with those in the B group (P < 0.05). These data suggest that branched-chain amino acid supplementation does not improve lean tissue accretion of low and normal birth weight pigs, despite a reduction in Sestrin2 expression in skeletal muscle of low birth weight pigs. The modest improvement in fractional growth rate of low birth weight pigs compared with their normal birth weight siblings was likely due to a more efficient dietary protein utilization.
Collapse
Affiliation(s)
- Joseph A Yonke
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA
| | - Kacie A Seymour
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA
| | - Samer W El-Kadi
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA.
| |
Collapse
|
42
|
Dangudubiyyam SV, Bosse B, Yadav P, Song R, Hofmann A, Mishra JS, Kumar S. Restoring Angiotensin Type 2 Receptor Function Reverses PFOS-Induced Vascular Hyper-Reactivity and Hypertension in Pregnancy. Int J Mol Sci 2023; 24:14180. [PMID: 37762482 PMCID: PMC10531530 DOI: 10.3390/ijms241814180] [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: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Perfluorooctane sulfonic acid (PFOS) exposure during pregnancy induces hypertension with decreased vasodilatory angiotensin type-2 receptor (AT2R) expression and impaired vascular reactivity and fetal weights. We hypothesized that AT2R activation restores the AT1R/AT2R balance and reverses gestational hypertension by improving vascular mechanisms. Pregnant Sprague-Dawley rats were exposed to PFOS through drinking water (50 μg/mL) from gestation day (GD) 4-20. Controls received drinking water with no detectable PFOS. Control and PFOS-exposed rats were treated with AT2R agonist Compound 21 (C21; 0.3 mg/kg/day, SC) from GD 15-20. In PFOS dams, blood pressure was higher, blood flow in the uterine artery was reduced, and C21 reversed these to control levels. C21 mitigated the heightened contraction response to Ang II and enhanced endothelium-dependent vasorelaxation in uterine arteries of PFOS dams. The observed vascular effects of C21 were correlated with reduced AT1R levels and increased AT2R and eNOS protein levels. C21 also increased plasma bradykinin production in PFOS dams and attenuated the fetoplacental growth restriction. These data suggest that C21 improves the PFOS-induced maternal vascular dysfunction and blood flow to the fetoplacental unit, providing preclinical evidence to support that AT2R activation may be an important target for preventing or treating PFOS-induced adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Sri Vidya Dangudubiyyam
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
| | - Bradley Bosse
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA;
| | - Pankaj Yadav
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
| | - Ruolin Song
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
| | - Alissa Hofmann
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
| | - Jay S. Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.V.D.); (P.Y.); (R.S.); (A.H.); (J.S.M.)
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA;
| |
Collapse
|
43
|
Daniele C, Farland LV, Park K, Schnatz PF, Shadyab AH, Stefanick ML, Wactawski-Wende J, Wild RA, Spracklen CN. Association of maternal birth weight and maternal preterm birth with subsequent risk for adverse reproductive outcomes: The Women's Health Initiative. Early Hum Dev 2023; 184:105839. [PMID: 37549575 PMCID: PMC10658641 DOI: 10.1016/j.earlhumdev.2023.105839] [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: 06/06/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Advancements in medical technology and pharmacologic interventions have drastically improved survival of infants born preterm and low birth weight, but knowledge regarding the long-term health impacts of these individuals is limited and inconsistent. AIM To investigate whether an individual's birthweight or history of being born preterm increases the risk of an adverse reproductive outcome. STUDY DESIGN Nested case-control study within the Women's Health Initiative. SUBJECTS 79,934 individuals who self-reported their personal birthweight category and/or preterm birth status. OUTCOMES MEASURES Self-reported pregnancy outcomes: subfertility, miscarriage, stillbirth, preeclampsia, gestational diabetes, gestational hypertension, preterm birth, low birthweight infant, high birthweight infant. Logistic regression models were used to estimate unadjusted and adjusted odds ratios (OR). RESULTS After adjustments, individuals reporting their birthweight <6lbs. were 20 % more likely to have a stillbirth or 70 % more likely to have a low birthweight infant and were less likely to have a full-term birth or high birthweight infant during their pregnancy. Individuals reporting a birthweight ≥10 lbs. were more likely to have a high birthweight infant (OR 3.49, 95 % CI 2.73-4.39) and less likely to have a low birthweight infant (OR 0.64, 95 % CI 0.47-0.82). Individuals born preterm were at increased risk for infertility, miscarriage, preeclampsia, gestational diabetes, and delivering a preterm or low birthweight infant. CONCLUSIONS As more individuals born preterm and/or low birthweight survive to adulthood, the incidence and prevalence of poor reproductive outcomes may increase. Women born at extremes of birthweight and prematurity may need to be monitored more closely during their own pregnancies.
Collapse
Affiliation(s)
- Christian Daniele
- Department of Biostatistics and Epidemiology, 715 North Pleasant Street, University of Massachusetts Amherst, Amherst, MA 01003, United States of America
| | - Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, 1295 N. Martin Ave, University of Arizona, Tucson, AZ 85724, United States of America
| | - Ki Park
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, 1600 SW Archer Road, University of Florida, Gainesville, FL 32610, United States of America
| | - Peter F Schnatz
- Departments of Obstetrics and Gynecology and Internal Medicine, Reading Hospital, Tower Health & Drexel University, 6th Avenue and Spruce Street, West Reading, PA 19611, United States of America
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, 9500 Gilman Drive #0725, La Jolla, CA 92093, United States of America
| | - Marcia L Stefanick
- Stanford Prevention Research Center, Department of Medicine, Stanford School of Medicine, Stanford University, 1265 Welch Road, Room X308, Stanford, CA 94305, United States of America
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, 174 Biomedical Education Building, Buffalo, NY 14214, United States of America
| | - Robert A Wild
- Department of Obstetrics and Gynecology, University of Oklahoma College of Medicine, 800 SL Young Blvd, Oklahoma City, OK 73104, United States of America; Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, 715 North Pleasant Street, University of Massachusetts Amherst, Amherst, MA 01003, United States of America.
| |
Collapse
|
44
|
Gualdoni GS, Barril C, Jacobo PV, Pacheco Rodríguez LN, Cebral E. Involvement of metalloproteinase and nitric oxide synthase/nitric oxide mechanisms in early decidual angiogenesis-vascularization of normal and experimental pathological mouse placenta related to maternal alcohol exposure. Front Cell Dev Biol 2023; 11:1207671. [PMID: 37670932 PMCID: PMC10476144 DOI: 10.3389/fcell.2023.1207671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023] Open
Abstract
Successful pregnancy for optimal fetal growth requires adequate early angiogenesis and remodeling of decidual spiral arterioles during placentation. Prior to the initiation of invasion and endothelial replacement by trophoblasts, interactions between decidual stromal cells and maternal leukocytes, such as uterine natural killer cells and macrophages, play crucial roles in the processes of early maternal vascularization, such as proliferation, apoptosis, migration, differentiation, and matrix and vessel remodeling. These placental angiogenic events are highly dependent on the coordination of several mechanisms at the early maternal-fetal interface, and one of them is the expression and activity of matrix metalloproteinases (MMPs) and endothelial nitric oxide synthases (NOSs). Inadequate balances of MMPs and nitric oxide (NO) are involved in several placentopathies and pregnancy complications. Since alcohol consumption during gestation can affect fetal growth associated with abnormal placental development, recently, we showed, in a mouse model, that perigestational alcohol consumption up to organogenesis induces fetal malformations related to deficient growth and vascular morphogenesis of the placenta at term. In this review, we summarize the current knowledge of the early processes of maternal vascularization that lead to the formation of the definitive placenta and the roles of angiogenic MMP and NOS/NO mechanisms during normal and altered early gestation in mice. Then, we propose hypothetical defective decidual cellular and MMP and NOS/NO mechanisms involved in abnormal decidual vascularization induced by perigestational alcohol consumption in an experimental mouse model. This review highlights the important roles of decidual cells and their MMP and NOS balances in the physiological and pathophysiological early maternal angiogenesis-vascularization during placentation in mice.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Cebral
- Laboratorio de Reproducción y Fisiología Materno-Embrionaria, Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Departamento de Biodiversidad y Biología Experimental (DBBE), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
45
|
Groene SG, Gremmen IJ, van Zwet EW, Roest AAW, Haak MC, van Klink JMM, Lopriore E, Heijmans BT, de Bruin C. Fetal growth restriction inhibits childhood growth despite catch-up in discordant identical twins: an observational cohort study. Eur J Endocrinol 2023; 189:183-189. [PMID: 37548178 DOI: 10.1093/ejendo/lvad103] [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: 03/12/2023] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVE Research suggests that postnatal catch-up growth after fetal growth restriction (FGR) occurs frequently. Yet, postnatal growth in singletons may be influenced by multiple factors. Identical twins with discordant prenatal growth, termed selective FGR (sFGR), can be regarded as a natural experiment eliminating these sources of bias. DESIGN Observational cohort study. METHODS Monochorionic twins with sFGR born between 2002 and 2017 (aged 3-17 years) were eligible. Growth measurements (height, weight, head circumference, and body mass index) were performed at follow-up. Detailed growth curves documented by a systematic primary care system in the Netherlands were collected. Measurements were converted to standard deviation scores (SDSs). A mixed-effects model was used to assess within-pair SDS difference and individual height SDS relative to target height SDS. RESULTS Forty-seven twin pairs (94 children) were included at a median age of 11 (interquartile range 8-13) years. At the last measurement, smaller twins at birth had a lower height SDS [-0.6 vs -0.3, P < .001, median difference 0.5 (95%CI 0.4-0.7)], lower weight SDS [-0.5 vs -0.1, P < .001, median difference 0.8 (95%CI 0.5-1.0)], and lower head circumference SDS [-0.5 vs 0.2, P < .001, median difference 0.8 (95%CI 0.6-0.9)] compared to larger twins. These differences persisted until the age of 17. Smaller twins showed rapid catch-up growth in the first 2 years and reached their target height range between 8 and 11 years. CONCLUSIONS Identical twins with discordant prenatal growth maintain a modest but significant difference in height, weight, and head circumference, indicating a persistent, inhibitory effect of an adverse intrauterine environment on childhood growth.
Collapse
Affiliation(s)
- Sophie G Groene
- Neonatology, Department of Pediatrics, Leiden University Medical Center, Willem-Alexander Children's Hospital, Leiden, The Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Irma J Gremmen
- Neonatology, Department of Pediatrics, Leiden University Medical Center, Willem-Alexander Children's Hospital, Leiden, The Netherlands
| | - Erik W van Zwet
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno A W Roest
- Pediatric Cardiology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique C Haak
- Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanine M M van Klink
- Neonatology, Department of Pediatrics, Leiden University Medical Center, Willem-Alexander Children's Hospital, Leiden, The Netherlands
| | - Enrico Lopriore
- Neonatology, Department of Pediatrics, Leiden University Medical Center, Willem-Alexander Children's Hospital, Leiden, The Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Christiaan de Bruin
- Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
46
|
Milà-Guasch M, Ramírez S, Llana SR, Fos-Domènech J, Dropmann LM, Pozo M, Eyre E, Gómez-Valadés AG, Obri A, Haddad-Tóvolli R, Claret M. Maternal emulsifier consumption programs offspring metabolic and neuropsychological health in mice. PLoS Biol 2023; 21:e3002171. [PMID: 37616199 PMCID: PMC10449393 DOI: 10.1371/journal.pbio.3002171] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 05/24/2023] [Indexed: 08/26/2023] Open
Abstract
Modern lifestyle is associated with a major consumption of ultra-processed foods (UPF) due to their practicality and palatability. The ingestion of emulsifiers, a main additive in UPFs, has been related to gut inflammation, microbiota dysbiosis, adiposity, and obesity. Maternal unbalanced nutritional habits during embryonic and perinatal stages perturb offspring's long-term metabolic health, thus increasing obesity and associated comorbidity risk. However, whether maternal emulsifier consumption influences developmental programming in the offspring remains unknown. Here, we show that, in mice, maternal consumption of dietary emulsifiers (1% carboxymethyl cellulose (CMC) and 1% P80 in drinking water), during gestation and lactation, perturbs the development of hypothalamic energy balance regulation centers of the progeny, leads to metabolic impairments, cognition deficits, and induces anxiety-like traits in a sex-specific manner. Our findings support the notion that maternal consumption of emulsifiers, common additives of UPFs, causes mild metabolic and neuropsychological malprogramming in the progeny. Our data call for nutritional advice during gestation.
Collapse
Affiliation(s)
- Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergio R. Llana
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lea Maria Dropmann
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alicia G. Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- School of Medicine, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
47
|
Oliveira MDS, Gonçalves FCLDSP, de Lira PIC, Filho SLVN, Eickmann SH, Lima MDC. Birthweight, postnatal growth and blood pressure in adolescents of low socioeconomic condition: a cohort study in Northeast Brazil. J Pediatr (Rio J) 2023; 99:391-398. [PMID: 36758624 PMCID: PMC10373154 DOI: 10.1016/j.jped.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
OBJECTIVE To verify the influence of low birth weight and rapid postnatal weight gain and associated factors on blood pressure in adolescence in a population of low socioeconomic status, considering that injuries occur in the perinatal period can be predictors of future metabolic changes and are still poorly explored. METHODS A cohort study was carried out with 208 adolescents, 78 born with low weight and 130 born with appropriate weight. The infants were followed up during the first six postnatal months and reassessed at 8 and 18 years of age. The independent variables were birthweight and postnatal weight gain. Rapid postnatal weight gain was defined when above 0.67 z score. The co-variables were sex, maternal height and family income at birth, nutritional status at eight years old, socioeconomic conditions, nutritional status, fat mass index, and physical activity level at 18 years. The outcome variable was blood pressure at 18 years old. The bivariate and multivariable logistic regression analysis were realized and p < 0,05 was considered significant. RESULTS The proportion of adolescents with elevated blood pressure was 37.5%. The multivariable logistic regression analysis showed the variables independently associated with a higher chance of elevated blood pressure in adolescence were rapid postnatal weight gain (OR = 2.74; 95% CI 1.22-6.14; p = 0.014), male sex (OR = 4.15; 95% CI 1.66-10.38; p = 0.002) and being physically active (OR = 2.70; 95% CI 1.08-6.74; p = 0.034). CONCLUSIONS The rapid postnatal weight gain was a predictor for elevated blood pressure in adolescence, independently of other factors.
Collapse
Affiliation(s)
| | | | - Pedro Israel Cabral de Lira
- Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, PE, Brazil; London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
| | | | - Sophie Helena Eickmann
- Universidade Federal de Pernambuco, Nutrição, Recife, PE, Brazil; Departamento Materno-Infantil, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Marilia de Carvalho Lima
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom; Departamento Materno-Infantil, Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
48
|
Oliveira WR, Rigo CP, Ferreira ARO, Ribeiro MVG, Perres MNC, Palma-Rigo K. Precocious evaluation of cardiovascular risk and its correlation with perinatal condition. AN ACAD BRAS CIENC 2023; 95:e20201702. [PMID: 37377255 DOI: 10.1590/0001-3765202320201702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/28/2021] [Indexed: 06/29/2023] Open
Abstract
The cardiovascular disease is the main cause of worldwide death. This profile is potentialized by the increased severity of infections in people with obesity, type 2 diabetes and hypertension. Children and adolescents are target groups for the prevention of non-communicable diseases. The Developmental Origins of Health and Disease concept points that perinatal conditions are an important risk factor to development of non-communicable disease in adulthood. In this context, the present review identifies perinatal factor that induces precocious cardiovascular risk factors, related with cardiometabolic syndrome. The low or high birth weight and caesarean delivery are risk factors that induce increased occurrence of cardiovascular risk biomarkers in children and adolescents, while the breast feeding or feeding with breast milk from the birth until two years-old is a protector strategy. Evaluation of perinatal conditions associated with precocious identification of cardiovascular risk factors in children and adolescents is an efficient strategy to prevent and control cardiovascular mortality; through interventions, as lifestyle changes during vulnerable windows of development, able to set up the risk to cardiometabolic disease.
Collapse
Affiliation(s)
- Wanderson R Oliveira
- Faculdade Adventista Paranaense, PR-317, Km 119, Gleba, Rua Paiçandu, Lote 80, Zona Rural, 87130-000 Ivatuba, PR, Brazil
| | - Cleusa P Rigo
- Centro Universitário Filadélfia, Rua Alagoas, 2050, Centro, 86010-520 Londrina, PR, Brazil
| | - Anna R O Ferreira
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Maiara V G Ribeiro
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Maria N C Perres
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Kesia Palma-Rigo
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
- Faculdade Adventista Paranaense, PR-317, Km 119, Gleba, Rua Paiçandu, Lote 80, Zona Rural, 87130-000 Ivatuba, PR, Brazil
| |
Collapse
|
49
|
Darby JRT, Williams GK, Cho SKS, Meakin AS, Holman SL, Quinn M, Wiese MD, Macgowan CK, Seed M, Morrison JL. Acute resveratrol exposure does not impact hemodynamics of the fetal sheep. Physiol Rep 2023; 11:e15749. [PMID: 37332034 PMCID: PMC10277215 DOI: 10.14814/phy2.15749] [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: 04/29/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Babies born growth restricted are at an increased risk of both poor short-and long-term outcomes. Current interventions to improve fetal growth are ineffective and do not lower the lifetime risk of poor health status. Maternal resveratrol (RSV) treatment increases uterine artery blood flow, fetal oxygenation, and fetal weight. However, studies suggest that diets high in polyphenols such as RSV may impair fetal hemodynamics. We aimed to characterize the effect of RSV on fetal hemodynamics to further assess its safety as an intervention strategy. Pregnant ewes underwent magnetic resonance imaging (MRI) scans to measure blood flow and oxygenation within the fetal circulation using phase contrast-MRI and T2 oximetry. Blood flow and oxygenation measures were performed in a basal state and then repeated while the fetus was exposed to RSV. Fetal blood pressure and heart rate were not different between states. RSV did not impact fetal oxygen delivery (DO2 ) or consumption (VO2 ). Blood flow and oxygen delivery throughout the major vessels of the fetal circulation were not different between basal and RSV states. As such, acute exposure of the fetus to RSV does not directly impact fetal hemodynamics. This strengthens the rationale for the use of RSV as an intervention strategy against fetal growth restriction.
Collapse
Affiliation(s)
- Jack R. T. Darby
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Georgia K. Williams
- Preclinical, Imaging and Research LaboratoriesSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Steven K. S. Cho
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
- Univeristy of Toronto and The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Ashley S. Meakin
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Stacey L. Holman
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Megan Quinn
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | | | - Mike Seed
- Univeristy of Toronto and The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Janna L. Morrison
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| |
Collapse
|
50
|
Saeed H, Lu YC, Andescavage N, Kapse K, Andersen NR, Lopez C, Quistorff J, Barnett S, Henderson D, Bulas D, Limperopoulos C. Influence of maternal psychological distress during COVID-19 pandemic on placental morphometry and texture. Sci Rep 2023; 13:7374. [PMID: 37164993 PMCID: PMC10172401 DOI: 10.1038/s41598-023-33343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/12/2023] [Indexed: 05/12/2023] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic has been accompanied by increased prenatal maternal distress (PMD). PMD is associated with adverse pregnancy outcomes which may be mediated by the placenta. However, the potential impact of the pandemic on in vivo placental development remains unknown. To examine the impact of the pandemic and PMD on in vivo structural placental development using advanced magnetic resonance imaging (MRI), acquired anatomic images of the placenta from 63 pregnant women without known COVID-19 exposure during the pandemic and 165 pre-pandemic controls. Measures of placental morphometry and texture were extracted. PMD was determined from validated questionnaires. Generalized estimating equations were utilized to compare differences in PMD placental features between COVID-era and pre-pandemic cohorts. Maternal stress and depression scores were significantly higher in the pandemic cohort. Placental volume, thickness, gray level kurtosis, skewness and run length non-uniformity were increased in the pandemic cohort, while placental elongation, mean gray level and long run emphasis were decreased. PMD was a mediator of the association between pandemic status and placental features. Altered in vivo placental structure during the pandemic suggests an underappreciated link between disturbances in maternal environment and perturbed placental development. The long-term impact on offspring is currently under investigation.
Collapse
Affiliation(s)
- Haleema Saeed
- Department of Obstetrics & Gynecology, MedStar Washington Hospital Center, Washington, DC, 20010, USA
| | - Yuan-Chiao Lu
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Nickie Andescavage
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Division of Neonatology, Children's National Hospital, Washington, DC, 20010, USA
| | - Kushal Kapse
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Nicole R Andersen
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Catherine Lopez
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Jessica Quistorff
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Scott Barnett
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Diedtra Henderson
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Dorothy Bulas
- Division of Radiology, Children's National Hospital, Washington, DC, 20010, USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA.
- Division of Radiology, Children's National Hospital, Washington, DC, 20010, USA.
| |
Collapse
|