1
|
Amanollahi R, Holman SL, Bertossa MR, Meakin AS, Clifton VL, Thornburg KL, McMillen IC, Wiese MD, Lock MC, Morrison JL. Elevated cortisol concentration in preterm sheep fetuses impacts heart development. Exp Physiol 2025. [PMID: 40296367 DOI: 10.1113/ep092506] [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: 12/11/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025]
Abstract
The prepartum rise in cortisol promotes cardiac development and maturation. Here, we investigated the impact of elevated circulating cortisol during mid-late gestation on cardiac growth and metabolism in fetal sheep. Saline or cortisol (2-3 mg in 4.4 mL/24 h) was infused into the fetal jugular vein from 109 to 116 days gestation (dG, term = 150 dG), and fetal heart tissue was collected at 116 dG. Glucocorticoid concentrations, gene and protein expression were measured in fetal left ventricle (LV) tissue. Intrafetal cortisol infusion increased cardiac cortisol concentration but downregulated the protein abundance of glucocorticoid receptor (GR) isoforms (GRα-A, GR-P, GR-A, GRα-D2 and GRα-D3). The gene and protein expression of markers of cardiac hyperplastic growth (IGF1, IGF-1R, TGFβ and AGT) were downregulated, while a protein marker of DNA replication (proliferating cell nuclear antigen) was upregulated by cortisol infusion. Cardiac protein and/or gene expression of complex I of the electron transport chain, SOD2, GLUT-4 (gene and protein), and phosphorylated IRS-1, were upregulated in response to elevated fetal cortisol concentration. Intrafetal cortisol infusion downregulated gene expression of PDK4, which mediates the metabolic switch from glucose to fatty acid metabolism. Cardiac expression of molecular markers involved in cardiovascular protection (SIRT-1, HO1, LAMP1 and SK1) were also downregulated in the cortisol group. In conclusion, these findings suggest that chronic cortisol exposure in preterm fetuses alters heart development, promoting cardiac maturation and potentially increasing the risk of cardiovascular disease later in life if these changes persist into adulthood.
Collapse
Affiliation(s)
- Reza Amanollahi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - 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
| | - Melanie R Bertossa
- 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
| | - Vicki L Clifton
- Pregnancy and Development Group, Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
| | - Kent L Thornburg
- Department of Medicine, Center for Developmental Health, Knight Cardiovascular Institute, Bob and Charlee Moore Institute of Nutrition and Wellness, Oregon Health & Science University, Portland, Oregon, USA
| | - I Caroline McMillen
- 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
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences University of South Australia, Adelaide, South Australia, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - 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
| |
Collapse
|
2
|
Sehgal A, Levins C, Yeomans E, Lu Z, Metz D. Proteinuria in preterm neonates: influence of fetal growth restriction. J Perinatol 2025:10.1038/s41372-025-02306-0. [PMID: 40253560 DOI: 10.1038/s41372-025-02306-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVE To compare proteinuria in preterm neonates with fetal growth restriction-small for gestational age (FGR-SGA) against equally preterm but appropriate for gestational age (AGA) neonates. STUDY DESIGN Prospective, observational cohort study. RESULTS Eighteen FGR-SGA neonates were compared with 18 AGA neonates (gestation; 29 ± 1 vs 29 ± 2 weeks, P = 0.8). Urine total protein (median [interquartile range]) in FGR-SGA was higher 370 [323, 573] vs 255 [193, 453] mg/L in AGA, P = 0.017 at first assessment (week one) and 565 [445, 743] vs 225 [135, 458] mg/L, P = 0.0011 at second assessment (week four). Urine protein creatinine ratio was 393 [250, 445] in FGR-SGA vs 227 [163, 367] mg/mmol in AGA, P = 0.029 at first assessment and 444 [368, 699] vs 240 [199, 411] mg/mmol, P = 0.0014 at second assessment. Mean blood pressure was higher in FGR-SGA group & directly correlated with proteinuria. CONCLUSIONS Increased proteinuria in FGR-SGA suggests reduced nephron endowment and hyper-filtration.
Collapse
Affiliation(s)
- Arvind Sehgal
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia.
- Department of Pediatrics, Monash University, Melbourne, VIC, Australia.
| | - Criona Levins
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC, Australia
| | - Emma Yeomans
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC, Australia
| | - Zhong Lu
- Department of Biochemistry, Monash Health, Melbourne, VIC, Australia
| | - David Metz
- Department of Pediatrics, Monash University, Melbourne, VIC, Australia
- Department of Nephrology, Royal Children's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Peers de Nieuwburgh M, Hunt M, Chandrasekaran P, Vincent TL, Hayes KB, Randazzo IR, Gunder M, De Bie FR, Colson A, Lu M, Wen H, Michki SN, Rychik J, Debiève F, Katzen J, Young LR, Davey MG, Flake AW, Gaynor JW, Frank DB. Chronic Hypoxia in an EXTrauterine Environment for Neonatal Development Impairs Lung Development. Am J Respir Cell Mol Biol 2025; 72:441-452. [PMID: 39453404 DOI: 10.1165/rcmb.2024-0012oc] [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: 01/11/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Severe fetal hypoxia poses a significant risk to lung development, resulting in severe postnatal complications. Existing chronic hypoxia animal models lack the ability to achieve pathologically reduced fetal oxygen without compromising animal development, placental blood flow, or maternal health. Using an established model of isolated chronic hypoxia involving the Extrauterine Environment for Neonatal Development, we are able to investigate the direct impact of fetal hypoxia on lung development. Oxygen delivery to preterm fetal lambs (105-110 d gestational age) delivered by cesarean section was reduced, and animals were supported using the Extrauterine Environment for Neonatal Development through the canalicular or saccular stage of lung development. Fetal lambs in hypoxic conditions showed significant growth restriction compared with their normoxic counterparts. We also observed modest aberrant vascular remodeling in the saccular group after hypoxic conditions, with decreased macrovessel numbers and microvascular endothelial cell numbers and increased peripheral vessel muscularization. In addition, fetal hypoxia resulted in enlarged distal airspaces and decreased septal wall volume. Moreover, there was a reduction in mature SFTPB (surfactant protein B) and processed SFTPC protein expression concomitant with a decrease in alveolar type 2 cell number. These findings demonstrate that maternally independent fetal hypoxia predominantly affects distal airway development, alveolar type 2 cell number, and surfactant production, with mild effects on the vasculature.
Collapse
Affiliation(s)
| | - Mallory Hunt
- Division of Cardiovascular Surgery, Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Tiffany L Vincent
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, and
- Service d'Obstétrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Sylvia N Michki
- Division of Cardiology
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Fréderic Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, and
- Service d'Obstétrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Jeremy Katzen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Lisa R Young
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
4
|
Divvela SSK, Gallorini M, Gellisch M, Patel GD, Saso L, Brand-Saberi B. Navigating redox imbalance: the role of oxidative stress in embryonic development and long-term health outcomes. Front Cell Dev Biol 2025; 13:1521336. [PMID: 40206404 PMCID: PMC11979171 DOI: 10.3389/fcell.2025.1521336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
Embryonic development is a complex process of concurrent events comprising cell proliferation, differentiation, morphogenesis, migration, and tissue remodeling. To cope with the demands arising from these developmental processes, cells increase their nutrient uptake, which subsequently increases their metabolic activity. Mitochondria play a key role in the maintenance of metabolism and production of reactive oxygen species (ROS) as a natural byproduct. Regulation of ROS by antioxidants is critical and tightly regulated during embryonic development, as dysregulation results in oxidative stress that damages essential cellular components such as DNA, proteins, and lipids, which are crucial for cellular maintenance and in extension development. However, during development, exposure to certain exogenous factors or damage to cellular components can result in an imbalance between ROS production and its neutralization by antioxidants, leading to detrimental effects on the developmental process. In this review article, we highlight the crucial role of redox homeostasis in normal development and how disruptions in redox balance may result in developmental defects.
Collapse
Affiliation(s)
| | - Marialucia Gallorini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Morris Gellisch
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Gaurav Deepak Patel
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
5
|
McGillick EV, Orgeig S, Allison BJ, Brain KL, Niu Y, Itani N, Skeffington KL, Kane AD, Herrera EA, Giussani DA, Morrison JL. Antenatal Vitamin C differentially affects lung development in normally grown and growth restricted sheep. Pediatr Res 2025:10.1038/s41390-025-03828-1. [PMID: 40000856 DOI: 10.1038/s41390-025-03828-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 12/07/2024] [Accepted: 12/31/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Chronic hypoxemia is a common cause of fetal growth restriction and can have significant effects on the developing fetal lung. Maternal antioxidant treatment in hypoxic pregnancy protects against offspring cardiovascular dysfunction. The effects of antenatal antioxidants on lung development in the chronically hypoxic growth restricted fetus is unknown. METHODS We investigated the effect of maternal daily Vitamin C (200 mg/kg i.v. vs. Saline) for a month in late gestation on molecular markers regulating lung maturation between normoxic normally grown and hypoxic growth-restricted fetal sheep. Chronic fetal hypoxia and fetal growth restriction were induced by exposure to maternal chronic hypoxia (10% O2 vs. Normoxia=21% O2) from 105-138 d gestation (term=145 d). RESULTS The data show a differential effect of antenatal Vitamin C treatment on regulation of genes involved in surfactant maturation, sodium movement and hypoxia signaling. Limited responsiveness to antenatal Vitamin C exposure in the lung of the hypoxic fetus, compared to responsiveness to antenatal Vitamin C in the normoxic fetus, suggests a maximal upregulation of the molecular signaling pathways in response to the chronic hypoxic insult alone. CONCLUSION We provide molecular insight into the heterogeneity of antenatal Vitamin C treatment on development of the normoxic and growth restricted hypoxic fetal lung. IMPACT The effect of maternal Vitamin C on molecular markers of lung maturation between normoxic normally grown and hypoxic growth restricted fetal sheep was unknown. We show a differential effect of Vitamin C with a greater increase in molecular markers of lung maturation in normoxic compared with hypoxic fetuses. Limited responsiveness in the hypoxic fetal lung is likely due to maximal upregulation by the hypoxic insult alone, thus added exposure to Vitamin C is unable to upregulate the system further. The work highlights the need to understand differential effects of antenatal interventions in healthy and complicated pregnancy, prior to clinical translation.
Collapse
Affiliation(s)
- Erin V McGillick
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Adelaide, Australia.
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Sandra Orgeig
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Beth J Allison
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Kirsty L Brain
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Youguo Niu
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Nozomi Itani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Katie L Skeffington
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Andrew D Kane
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Emilio A Herrera
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Adelaide, Australia.
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
6
|
McGillick EV, Orgeig S, Allison BJ, Brain KL, Bertossa MR, Holman SL, Meakin AS, Wiese MD, Niu Y, Itani N, Skeffington KL, Beck C, Botting-Lawford KJ, Morrison JL, Giussani DA. Chronic fetal hypoxia and antenatal Vitamin C exposure differentially regulate molecular signalling in the lung of female lambs in early adulthood. Front Physiol 2025; 15:1488152. [PMID: 39882327 PMCID: PMC11775154 DOI: 10.3389/fphys.2024.1488152] [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: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Chronic fetal hypoxia is commonly associated with fetal growth restriction and can predispose to respiratory disease at birth and in later life. Antenatal antioxidant treatment has been investigated to overcome the effects of oxidative stress in utero to improve respiratory outcomes. We aimed to determine if the effects of chronic fetal hypoxia and antenatal antioxidant administration persist in the lung in early adulthood. Methods Chronically catheterised pregnant sheep were exposed to normoxia (N; n = 20) or hypoxia (H; n = 18; 10% O2) ± maternal daily i. v. saline (N = 11; H = 8) or Vitamin C (VC; NVC = 9; HVC = 10) from 105 to 138 days (term, ∼145 days). Lungs were collected from female lambs 9 months after birth (early adulthood). Lung tissue expression of genes and proteins regulating oxidative stress, mitochondrial function, hypoxia signalling, glucocorticoid signalling, surfactant maturation, inflammation and airway remodelling were measured. Results Chronic fetal hypoxia upregulated lung expression of markers of prooxidant, surfactant lipid transport and airway remodelling pathways in early adulthood. Antenatal Vitamin C normalized prooxidant and airway remodelling markers, increased endogenous antioxidant, vasodilator and inflammatory markers, and altered regulation of hypoxia signalling and glucocorticoid availability. Conclusion There are differential effects of antenatal Vitamin C on molecular markers in the lungs of female lambs from normoxic and hypoxic pregnancy in early adulthood.
Collapse
Affiliation(s)
- Erin V. McGillick
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, University of South Australia, Adelaide, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Sandra Orgeig
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Beth J. Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kirsty L. Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Melanie R. Bertossa
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, University of South Australia, Adelaide, Australia
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, University of South Australia, Adelaide, Australia
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, University of South Australia, Adelaide, Australia
| | - Michael D. Wiese
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Nozomi Itani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Katie L. Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Christian Beck
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | | | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, University of South Australia, Adelaide, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Dino A. Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
7
|
van Aswegen M, Szabo A, Currie JJ, Stack SH, West KL, Hofmann N, Christiansen F, Bejder L. Energetic cost of gestation and prenatal growth in humpback whales. J Physiol 2025; 603:529-550. [PMID: 39661448 DOI: 10.1113/jp287304] [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: 07/12/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
Improving our understanding of energy allocation in reproduction is key for accurately parameterizing bioenergetic models to assess population responses to environmental perturbations and anthropogenic disturbance. We quantified the energetic cost of gestation in humpback whales (Megaptera novaeangliae) using historical whaling records, non-invasive unoccupied aerial system (UAS) photogrammetry and post mortem tissue samples. First, we estimated relative birth size using body length measurements of 678 mother-fetus pairs from historical whaling records and 987 mother-calf pairs measured in situ using UAS-photogrammetry. The total energetic cost of gestation includes fetal growth (FG), heat increment of gestation and placental tissue development. FG was modelled from conception to birth, with fetal volume and mass estimated using the volume-to-length relationship of perinatal calves and published humpback whale tissue composition estimates. Tissue-specific energy content was quantified using post mortem bone, muscle, viscera and blubber samples from a neonatal humpback whale. Placental tissue development was estimated using humpback whale placental tissue and published equations. Relative birth length was found to be 33.75% (95% CI: 32.10-34.61) of maternal length. FG rates and absolute birth size increased with maternal length, with exponential growth in fetal length, volume and mass resulting in minimal energetic costs over the first two quadmesters (0.01-1.08%) before increasing significantly in the final quadmester (98.92%). Gestational heat constituted the greatest energetic cost (90.42-94.95%), followed by fetal (4.58-7.76%) and placental (0.37-1.83%) tissue growth. Our findings highlight the energetic costs endured by capital breeding females preceding parturition, with the most substantial energetic costs of gestation coinciding with migration and fasting. KEY POINTS: We quantified the energetic cost of gestation using body length measurements of mother-fetus pairs from historical whaling records, length estimates of mother-calf pairs measured in situ using aerial photogrammetry and post mortem tissue samples. Fetal growth rates and birth size increased with maternal length, with fetal length, volume and mass increasing exponentially over gestation. Energetic costs over the first two quadmesters were negligible (0.01-1.08%) before increasing significantly in the final quadmester (98.92%). Though larger females incur nearly twice the energetic cost of smaller females, they are likely buffered by greater absolute energy reserves, suggesting smaller females may be less resilient to perturbations in energy balance. We demonstrate the significant energetic costs incurred by pregnant humpback whales, with most of the energetic expenditure occurring over the final 100 days of gestation. Late-pregnant females are, therefore, particularly vulnerable to disruptions in energy balance, given periods of greatest energetic stress coincide with fasting and migration.
Collapse
Affiliation(s)
- Martin van Aswegen
- Marine Mammal Research Program, Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, USA
- Alaska Whale Foundation, Petersburg, AK, USA
| | - Andy Szabo
- Alaska Whale Foundation, Petersburg, AK, USA
- Hawai'i Institute of Marine Biology, University of Hawai'i at Manoa, Kāne'ohe, Hawai'i, USA
| | - Jens J Currie
- Marine Mammal Research Program, Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, USA
- Pacific Whale Foundation, Maui, HI, USA
| | - Stephanie H Stack
- Pacific Whale Foundation, Maui, HI, USA
- Southern Ocean Persistent Organic Pollutants Program, School of Environment and Science, Griffith University, Queensland, Australia
| | - Kristi L West
- Health and Stranding Lab, Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, USA
- Health and Stranding Lab, College of Tropical Agriculture and Human Resources, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Nicholas Hofmann
- Health and Stranding Lab, Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, USA
| | - Fredrik Christiansen
- Marine Mammal Research, Department of Ecoscience, Aarhus University, Roskilde, Denmark
| | - Lars Bejder
- Marine Mammal Research Program, Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, USA
- Zoophysiology, Department of Bioscience, Aarhus University, Aarhus, Denmark
| |
Collapse
|
8
|
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
|
9
|
Zi Y, Qin Y, Ma C, Qiao Y, Xu X, Yang Y, He Q, Li M, Liu Y, Gao F. Transcriptome analysis reveals hepatic disordered lipid metabolism, lipotoxic injury, and abnormal development in IUGR sheep fetuses due to maternal undernutrition during late pregnancy. Theriogenology 2024; 226:350-362. [PMID: 38968678 DOI: 10.1016/j.theriogenology.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 06/19/2024] [Accepted: 06/23/2024] [Indexed: 07/07/2024]
Abstract
Although lipid metabolism in fetal livers under intrauterine growth restriction (IUGR) conditions has been widely studied, the implications of maternal undernutrition on fetal hepatic lipid metabolism, lipotoxic injury, and abnormal development remain largely unknown. Therefore, this study investigated the effects of maternal undernutrition on disordered hepatic lipid metabolism, lipotoxic injury, and abnormal development in IUGR sheep fetuses using transcriptome analysis. Seventeen singleton ewes were randomly divided into three groups on day 90 of pregnancy: a control group (CG; 0.63 MJ metabolic energy/body weight (ME/BW)0.75/day, n = 5), maternal undernutrition group 1 (MU1; 0.33 MJ ME/BW0.75/day, n = 6), and maternal undernutrition group 2 (MU2; 0.20 MJ ME/BW0.75/day, n = 6). The fetuses were euthanized and recovered on day 130 of pregnancy. The levels of free fatty acids (FFA) in maternal blood (P < 0.01), fetal blood (P < 0.01), and fetal livers (P < 0.05) were increased in the MU1 and MU2 groups, but fetal hepatic triglyceride (TG) levels in the MU2 group (P < 0.01) and β-hydroxybutyrate levels in the MU1 and MU2 groups (P < 0.01) were decreased compared to the CG. Severe inflammatory cell infiltration and increased non-alcoholic fatty liver disease activity scores were observed in MU1 and MU2 fetuses (P < 0.01). Progressive deposition of fetal hepatic reticular fibers and collagen fibers in the fetal livers of the MU1 and MU2 groups and significant hepatic fibrosis were observed in the MU2 fetuses (P < 0.05). Gene set enrichment analysis showed that genes involved in lipid accumulation and FFA beta oxidation were downregulated in both MU groups compared to those in the controls. The fetal liver mRNA expression of the β-oxidation regulator, acetyl-CoA acetyltransferase 1, and the TCA regulator, isocitrate dehydrogenase were reduced in MU1 (P < 0.05) and MU2 (P < 0.01) fetuses, and downregulated mRNA expression of long chain fatty acid CoA ligase 1 (P < 0.05) and glycerol-3-phosphate acyltransferase (P < 0.01) was observed in MU2 fetuses. Differentially expressed genes (DEGs) in MU1 versus CG (360 DEGs) and MU2 versus CG (746 DEGs) were identified using RNA sequencing. Bioinformatics analyses of the 231 intersecting DEGs between MU1 versus CG and MU2 versus CG indicated that neutrophil extracellular traps (NETs) were induced and played a central role in fetal hepatic injury in IUGR sheep. Increased maternal blood myeloperoxidase (MPO) levels (P < 0.01), NE (Elane)-positive areas in fetal liver sections (P < 0.05), and fetal liver MPO protein expression (P < 0.01) were found in the MU1 and MU2 groups; however, MPO levels were reduced in the fetal membrane (P < 0.01) and fetal blood (P < 0.05) in the MU1 group, and in the maternal-fetal placenta and fetal blood in the MU2 group (P < 0.01). Analysis of gene expression trends in the intersecting DEGs between MU1 versus CG (129 DEGs) and MU2 versus CG (515 DEGs) further revealed that 30 hub genes were essential regulators of the G2/M cell cycle, all of which were associated with hepatocellular carcinoma. G0/G1 phase cells of the fetal liver were reduced in the MU1 (P < 0.05) and MU2 (P < 0.01) groups, whereas G2/M phase cells were elevated in the MU1 and MU2 groups (P < 0.01). The representatives of upregulated hub genes and fetal liver protein expression of maternal embryonic leucine zipper kinase and protein regulator of cytokinesis 1 were progressively enhanced in the MU1 and MU2 groups (P < 0.01), and topoisomerase II alpha protein expression in the MU2 group (P < 0.05), as expected. These results indicate that FFA overload, severe lipotoxic injury, and NETs were induced, and disease-promoting regulators of the G2/M cell cycle were upregulated in the fetal liver of IUGR sheep. These findings provide new insights into the pathogenesis of impaired hepatic lipid metabolism and abnormal development and the molecular origin of post-natal liver disease in IUGR due to maternal undernutrition. This information can support the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yang Zi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China; Shenzheng Institute of Advanced Technology, Chinese Academy of Sciences, Shenzheng, China
| | - Yulong Qin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Chi Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yina Qiao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Xiaoyi Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yilin Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Qiuyue He
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Mingyue Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yingchun Liu
- College of Life Science, Inner Mongolia Key Laboratory of Biomanufacturing, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Feng Gao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Gallagher LT, Bardill J, Sucharov CC, Wright CJ, Karimpour-Fard A, Zarate M, Breckenfelder C, Liechty KW, Derderian SC. Dysregulation of miRNA-mRNA expression in fetal growth restriction in a caloric restricted mouse model. Sci Rep 2024; 14:5579. [PMID: 38448721 PMCID: PMC10918062 DOI: 10.1038/s41598-024-56155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Fetal growth restriction (FGR) is associated with aberrant placentation and accounts for a significant proportion of perinatal deaths. microRNAs have been shown to be dysregulated in FGR. The purpose of this study was to determine microRNA-regulated molecular pathways altered using a caloric restricted mouse model of FGR. Pregnant mice were subjected to a 50% caloric restricted diet beginning at E9. At E18.5, RNA sequencing of placental tissue was performed to identify differences in gene expression between caloric restricted and control placentas. Significant differences in gene expression between caloric restricted and control placentas were observed in 228 of the 1546 (14.7%) microRNAs. Functional analysis of microRNA-mRNA interactions demonstrated enrichment of several biological pathways with oxidative stress, apoptosis, and autophagy pathways upregulated and angiogenesis and signal transduction pathways downregulated. Ingenuity pathway analysis also suggested that ID1 signaling, a pathway integral for trophoblast differentiation, is also dysregulated in caloric restricted placentas. Thus, a maternal caloric restriction mouse model of FGR results in aberrant microRNA-regulated molecular pathways associated with angiogenesis, oxidative stress, signal transduction, apoptosis, and cell differentiation. As several of these pathways are dysregulated in human FGR, our findings suggest that this model may provide an excellent means to study placental microRNA derangements seen in FGR.
Collapse
Affiliation(s)
- Lauren T Gallagher
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - James Bardill
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Anis Karimpour-Fard
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Miguel Zarate
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Courtney Breckenfelder
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Kenneth W Liechty
- Division of Pediatric Surgery, University of Arizona College of Medicine, Tucson, AZ, 85721, USA
| | - S Christopher Derderian
- Colorado Fetal Care Center, Children's Hospital Colorado, University of Colorado, 13123 E 16th Ave, Aurora, CO, 80045, USA.
- Division of Pediatric Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Estrella CAS, Gatford KL, Xiang R, Javadmanesh A, Ghanipoor-Samami M, Nattrass GS, Shuaib E, McAllister MM, Beckman I, Thomsen DA, Clifton VL, Owens JA, Roberts CT, Hiendleder S, Kind KL. Asymmetric growth-limiting development of the female conceptus. Front Endocrinol (Lausanne) 2024; 14:1306513. [PMID: 38362586 PMCID: PMC10867182 DOI: 10.3389/fendo.2023.1306513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/08/2023] [Indexed: 02/17/2024] Open
Abstract
Introduction Sex differences in prenatal growth may contribute to sex-dependent programming effects on postnatal phenotype. Methods We integrated for the first time phenotypic, histomorphological, clinico-chemical, endocrine and gene expression analyses in a single species, the bovine conceptus at mid-gestation. Results We demonstrate that by mid-gestation, before the onset of accelerated growth, the female conceptus displays asymmetric lower growth compared to males. Female fetuses were smaller with lower ponderal index and organ weights than males. However, their brain:body weight, brain:liver weight and heart:body weight ratios were higher than in males, indicating brain and heart 'sparing'. The female placenta weighed less and had lower volumes of trophoblast and fetal connective tissue than the male placenta. Female umbilical cord vessel diameters were smaller, and female-specific relationships of body weight and brain:liver weight ratios with cord vessel diameters indicated that the umbilico-placental vascular system creates a growth-limiting environment where blood flow is redistributed to protect brain and heart growth. Clinico-chemical indicators of liver perfusion support this female-specific growth-limiting phenotype, while lower insulin-like growth factor 2 (IGF2) gene expression in brain and heart, and lower circulating IGF2, implicate female-specific modulation of key endocrine mediators by nutrient supply. Conclusion This mode of female development may increase resilience to environmental perturbations in utero and contribute to sex-bias in programming outcomes including susceptibility to non-communicable diseases.
Collapse
Affiliation(s)
- Consuelo Amor S. Estrella
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Kathryn L. Gatford
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ruidong Xiang
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Ali Javadmanesh
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Mani Ghanipoor-Samami
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Greg S. Nattrass
- South Australian Research and Development Institute, Livestock Systems, Roseworthy, SA, Australia
| | - Entesar Shuaib
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Milton M. McAllister
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Ian Beckman
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Dana A. Thomsen
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Vicki L. Clifton
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Julie A. Owens
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Deakin University, Geelong, VIC, Australia
| | - Claire T. Roberts
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Flinders University, College of Medicine and Public Health, Adelaide, SA, Australia
| | - Stefan Hiendleder
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Karen L. Kind
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
14
|
Abe T, Sarentonglaga B, Nagao Y. Advancements in medical research using fetal sheep: Implications for human health and treatment methods. Anim Sci J 2024; 95:e13945. [PMID: 38651196 DOI: 10.1111/asj.13945] [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: 01/24/2024] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Sheep are typically considered as industrial animals that provide wool and meals. However, they play a significant role in medical research in addition to their conventional use. Notably, sheep fetuses are resistant to surgical invasions and can endure numerous manipulations, such as needle puncture and cell transplantation, and surgical operations requiring exposure beyond the uterus. Based on these distinguishing characteristics, we established a chimeric sheep model capable of producing human/monkey pluripotent cell-derived blood cells via the fetal liver. Furthermore, sheep have become crucial as human fetal models, acting as platforms for developing and improving techniques for intrauterine surgery to address congenital disorders and clarifying the complex pharmacokinetic interactions between mothers and their fetuses. This study emphasizes the significant contributions of fetal sheep to advancing human disease understanding and treatment strategies, highlighting their unique characteristics that are not present in other animals.
Collapse
Affiliation(s)
- Tomoyuki Abe
- Open Science Laboratory, Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan
| | | | - Yoshikazu Nagao
- Department of Agriculture, Utsunomiya University, Tochigi, Japan
| |
Collapse
|
15
|
Gregorovicova M, Lashkarinia SS, Yap CH, Tomek V, Sedmera D. Hemodynamics During Development and Postnatal Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:201-226. [PMID: 38884713 DOI: 10.1007/978-3-031-44087-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
A well-developed heart is essential for embryonic survival. There are constant interactions between cardiac tissue motion and blood flow, which determine the heart shape itself. Hemodynamic forces are a powerful stimulus for cardiac growth and differentiation. Therefore, it is particularly interesting to investigate how the blood flows through the heart and how hemodynamics is linked to a particular species and its development, including human. The appropriate patterns and magnitude of hemodynamic stresses are necessary for the proper formation of cardiac structures, and hemodynamic perturbations have been found to cause malformations via identifiable mechanobiological molecular pathways. There are significant differences in cardiac hemodynamics among vertebrate species, which go hand in hand with the presence of specific anatomical structures. However, strong similarities during development suggest a common pattern for cardiac hemodynamics in human adults. In the human fetal heart, hemodynamic abnormalities during gestation are known to progress to congenital heart malformations by birth. In this chapter, we discuss the current state of the knowledge of the prenatal cardiac hemodynamics, as discovered through small and large animal models, as well as from clinical investigations, with parallels gathered from the poikilotherm vertebrates that emulate some hemodynamically significant human congenital heart diseases.
Collapse
Affiliation(s)
- Martina Gregorovicova
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Choon Hwai Yap
- Department of Bioengineering, Imperial College, London, UK
| | - Viktor Tomek
- Pediatric Cardiology, Motol University Hospital, Prague, Czech Republic
| | - David Sedmera
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
16
|
Motta G, Thangaraj SV, Padmanabhan V. Developmental Programming: Impact of Prenatal Exposure to Bisphenol A on Senescence and Circadian Mediators in the Liver of Sheep. TOXICS 2023; 12:15. [PMID: 38250971 PMCID: PMC10818936 DOI: 10.3390/toxics12010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
Prenatal exposure to endocrine disruptors such as bisphenol A (BPA) plays a critical role in the developmental programming of liver dysfunction that is characteristic of nonalcoholic fatty liver disease (NAFLD). Circadian and aging processes have been implicated in the pathogenesis of NAFLD. We hypothesized that the prenatal BPA-induced fatty-liver phenotype of female sheep is associated with premature hepatic senescence and disruption in circadian clock genes. The expression of circadian rhythm and aging-associated genes, along with other markers of senescence such as telomere length, mitochondrial DNA copy number, and lipofuscin accumulation, were evaluated in the liver tissue of control and prenatal BPA groups. Prenatal BPA exposure significantly elevated the expression of aging-associated genes GLB1 and CISD2 and induced large magnitude differences in the expression of other aging genes-APOE, HGF, KLOTHO, and the clock genes PER2 and CLOCK-in the liver; the other senescence markers remained unaffected. Prenatal BPA-programmed aging-related transcriptional changes in the liver may contribute to pathological changes in liver function, elucidating the involvement of aging genes in the pathogenesis of liver steatosis.
Collapse
Affiliation(s)
| | | | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48105, USA; (G.M.); (S.V.T.)
| |
Collapse
|
17
|
Ashley RL, Trigo EM, Ervin JM. Placental insufficiency and heavier placentas in sheep after suppressing CXCL12/CXCR4 signaling during implantation†. Biol Reprod 2023; 109:982-993. [PMID: 37724932 PMCID: PMC10724462 DOI: 10.1093/biolre/ioad122] [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: 06/08/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
During implantation, trophoblast cell invasion and differentiation is predominantly important to achieving proper placental formation and embryonic development. The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) working through its receptor C-X-C motif chemokine receptor 4 (CXCR4) is implicated in implantation and placentation but precise roles of this axis are unclear. Suppressing CXCL12/CXCR4 signaling at the fetal-maternal interface in sheep reduces trophoblast invasion, disrupts uterine remodeling, and diminishes placental vascularization. We hypothesize these negative impacts during implantation will manifest as compromised fetal and placental growth at midgestation. To test, on day 12 postbreeding, osmotic pumps were surgically installed in 30 ewes and delivered intrauterine CXCR4 inhibitor or saline for 7 or 14 days. On day 90, fetal/maternal tissues were collected, measured, weighed, and maternal (caruncle) and fetal (cotyledon) placenta components separated and analyzed. The objectives were to determine if (i) suppressing CXCL12/CXCR4 during implantation results in reduced fetal and placental growth and development and (ii) if varying the amount of time CXCL12/CXCR4 is suppressed impacts fetal/placental development. Fetal weights were similar; however greater placental weight and placentome numbers occurred when CXCL12/CXCR4 was suppressed for 14 days. In caruncles, greater abundance of fibroblast growth factor 2, vascular endothelial growth factor A, vascular endothelial growth factor A receptor 1 (FLT-1), and placental growth factor were observed after suppressing CXCL12/CXCR4. Similar results occurred in cotyledons except less vascular endothelial growth factor in 7 day group and less fibroblast growth factor in 14 day group. Our data underscore the importance of CXCL12/CXCR4 signaling during placentation and provide strong evidence that altering CXCL12-mediated signaling induces enduring placental effects manifesting later in gestation.
Collapse
Affiliation(s)
- Ryan L Ashley
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Elisa M Trigo
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Jacqueline M Ervin
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
18
|
Saini BS, Ducas R, Darby JRT, Marini D, Sun L, Macgowan CK, Windrim R, Kingdom JC, Wald RM, Morrison JL, Seed M. Feasibility of MRI assessment of maternal-fetal oxygen transport and consumption relative to maternal position in healthy late gestational pregnancies. J Physiol 2023; 601:5413-5436. [PMID: 37906114 DOI: 10.1113/jp285097] [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: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
Late gestational supine positioning reduces maternal cardiac output due to inferior vena caval (IVC) compression, despite increased collateral venous return. However, little is known about the impact of maternal position on oxygen (O2 ) delivery and consumption of the gravid uterus, fetus, placenta and lower limbs. We studied the effects of maternal positioning on these parameters in 20 healthy pregnant subjects at 36 ± 2 weeks using magnetic resonance imaging (MRI); a follow-up MRI was performed 6-months postpartum (n = 16/20). MRI techniques included phase-contrast and T1/T2 relaxometry for blood flow and oximetry imaging, respectively. O2 transport was measured in the following vessels (bilateral where appropriate): maternal abdominal descending aorta (DAoabdo ), IVC, ovarian, paraspinal veins (PSV), uterine artery (UtA) and external iliacs, and umbilical. Maternal cardiac output was measured by summing DAothoracic and superior vena cava flows. Supine mothers (n = 6) had lower cardiac output and O2 delivery in the DAoabdo , UtA and external iliac arteries, and higher PSV flow than those in either the left (n = 8) or right (n = 6) lateral positions during MRI. However, O2 consumption in the gravid uterus, fetus, placenta and lower limbs was unaffected by maternal positioning. The ratio of IVC/PSV flow decreased in supine mothers while ovarian venous flow and O2 saturation were unaltered, suggesting a major route of pelvic venous return unaffected by maternal position. Placental-fetal O2 transport and consumption were similar between left and right lateral maternal positions. In comparison to non-pregnant findings, DAoabdo and UtA O2 delivery and pelvic O2 consumption increased, while lower-limb consumption remained constant , despite reduced external iliac artery O2 delivery in late gestation. KEY POINTS: Though sleeping supine during the third trimester is associated with an increased risk of antepartum stillbirth, the underlying biological mechanisms are not fully understood. Maternal cardiac output and uteroplacental flow are reduced in supine mothers due to inferior vena caval compression from the weight of the gravid uterus. This MRI study provides a comprehensive circulatory assessment, demonstrating reduced maternal cardiac output and O2 delivery (uteroplacental, lower body) in supine compared to lateral positioning; however, O2 consumption (gravid uterus, fetus, placenta, lower limbs) was preserved. Unlike other mammalian species, the ovarian veins conduct substantial venous return from the human pregnant uterus that is unaffected by maternal positioning. Lumbar paraspinal venous flow increased in supine mothers. These observations may have important considerations during major pelvic surgery in pregnancy (i.e. placenta percreta). Future studies should address the importance of maternal positioning as a potential tool to deliver improved perinatal outcomes in pregnancies with compromised uteroplacental O2 delivery.
Collapse
Affiliation(s)
- Brahmdeep S Saini
- Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Robin Ducas
- Department of Internal Medicine, Section of Cardiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - 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
| | - Davide Marini
- Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Liqun Sun
- Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christopher K Macgowan
- Translational Medicine, 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
| | - Rory Windrim
- Department of Obstetrics and Gynaecology, Maternal-Fetal Medicine Division, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John C Kingdom
- Department of Obstetrics and Gynaecology, Maternal-Fetal Medicine Division, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel M Wald
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, 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, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mike Seed
- Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Evans NP, Bellingham M, Elcombe CS, Ghasemzadeh-Hasankolaei M, Lea RG, Sinclair KD, Padmanabhan V. Sexually dimorphic impact of preconceptional and gestational exposure to a real-life environmental chemical mixture (biosolids) on offspring growth dynamics and puberty in sheep. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104257. [PMID: 37659607 DOI: 10.1016/j.etap.2023.104257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023]
Abstract
Humans are ubiquitously exposed to complex mixtures of environmental chemicals (ECs). This study characterised changes in post-natal and peripubertal growth, and the activation of the reproductive axis, in male and female offspring of sheep exposed to a translationally relevant EC mixture (in biosolids), during pregnancy. Birthweight in both sexes was unaffected by gestational biosolids exposure. In contrast to females (unaffected), bodyweight in biosolids males was significantly lower than controls across the peripubertal period, however, they exhibited catch-up growth eventually surpassing controls. Despite weighing less, testosterone concentrations were elevated earlier, indicative of early puberty in the biosolids males. This contrasted with females in which the mean date of puberty (first progesterone cycle) was delayed. These results demonstrate that developmental EC-mixture exposure has sexually dimorphic effects on growth, puberty and the relationship between body size and puberty. Such programmed metabolic/reproductive effects could have significant impacts on human health and wellbeing.
Collapse
Affiliation(s)
- Neil P Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| | - Michelle Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Christopher S Elcombe
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Mohammad Ghasemzadeh-Hasankolaei
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Richard G Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Kevin D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | | |
Collapse
|
20
|
Schofield LG, Kahl RGS, Rodrigues SL, Fisher JJ, Endacott SK, Delforce SJ, Lumbers ER, Martin JH, Pringle KG. Placental deficiency of the (pro)renin receptor ((P)RR) reduces placental development and functional capacity. Front Cell Dev Biol 2023; 11:1212898. [PMID: 37588662 PMCID: PMC10427116 DOI: 10.3389/fcell.2023.1212898] [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: 04/27/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023] Open
Abstract
The (pro)renin receptor ((P)RR; also known as ATP6AP2) is a multifunctional receptor. The (P)RR activates the tissue renin-angiotensin system (RAS) and is also involved in regulating integral intracellular pathways such as V-ATPase and Wnt/β-catenin signalling. Given this, the (P)RR may be associated with essential pathways in placentation, however its role within the context of pregnancy remains poorly characterised. The first trimester/extravillous trophoblast cell line, HTR-8/SVneo, underwent an siRNA knockdown where they were incubated for 24 h with a negative control siRNA or siRNA targeting ATP6AP2 mRNA. xCELLigence real-time cell analysis was performed to assess the effect of ATP6AP2 mRNA knockdown on HTR-8/SVneo cell proliferation, migration, and invasion. In subsequent experiments, GFP-encoding lentiviral packaged gene-constructs were used to knockdown (P)RR expression in the trophectoderm of C57/BL6/CBA-F1 mouse blastocysts. Blastocysts were incubated for 6 h with vehicle (no-virus), control virus (non-targeting shRNA and GFP), or (P)RR-knockdown virus ((P)RR shRNA and GFP) before transfer into recipient pseudo-pregnant Swiss CD1 female mice. Fetal and placental tissues were collected and assessed at embryonic age (EA) 10 and 18. (P)RR levels were measured in the labyrinth zone of day 18 placentae and stereological Merz grid analysis was performed to determine the volumetric distribution of trophoblasts, fetal capillaries, and the maternal blood space. We showed that a reduction of ATP6AP2 expression in HTR-8/SVneo cells in vitro, impaired trophoblast proliferation, migration, and invasion. In vivo, decreasing placental labyrinth (P)RR expression adversely effected placental physiology, decreasing placental trophoblast number and total surface area available for exchange, while also increasing maternal blood space. Additionally, decreased (P)RR affected placental efficacy evident by the reduced fetal-placental weight ratio. Our study shows that the (P)RR is necessary for appropriate placental development and function.
Collapse
Affiliation(s)
- Lachlan G. Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Richard G. S. Kahl
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Samantha L. Rodrigues
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Joshua J. Fisher
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Saije K. Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sarah J. Delforce
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Eugenie R. Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jacinta H. Martin
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Kirsty G. Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
21
|
Lock MC, Botting KJ, Allison BJ, Niu Y, Ford SG, Murphy MP, Orgeig S, Giussani DA, Morrison JL. MitoQ as an antenatal antioxidant treatment improves markers of lung maturation in healthy and hypoxic pregnancy. J Physiol 2023; 601:3647-3665. [PMID: 37467062 PMCID: PMC10952154 DOI: 10.1113/jp284786] [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: 04/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023] Open
Abstract
Chronic fetal hypoxaemia is a common pregnancy complication that increases the risk of infants experiencing respiratory complications at birth. In turn, chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in animal models of hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. However, whether antenatal antioxidant therapy confers any benefit on lung development in complicated pregnancies has not yet been investigated. Here, we tested the hypothesis that maternal antenatal treatment with MitoQ will protect the developing lung in hypoxic pregnancy in sheep, a species with similar fetal lung developmental milestones as humans. Maternal treatment with MitoQ during late gestation promoted fetal pulmonary surfactant maturation and an increase in the expression of lung mitochondrial complexes III and V independent of oxygenation. Maternal treatment with MitoQ in hypoxic pregnancy also increased the expression of genes regulating liquid reabsorption in the fetal lung. These data support the hypothesis tested and suggest that MitoQ as an antenatal targeted antioxidant treatment may improve lung maturation in the late gestation fetus. KEY POINTS: Chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. MitoQ is a targeted antioxidant that uses the cell and the mitochondrial membrane potential to accumulate within the mitochondria. Treatment of healthy or hypoxic pregnancy with MitoQ, increases the expression of key molecules involved in surfactant maturation, lung liquid reabsorption and in mitochondrial proteins driving ATP synthesis in the fetal sheep lung. There were no detrimental effects of MitoQ treatment alone on the molecular components measured in the present study, suggesting that maternal antioxidant treatment has no effect on other components of normal maturation of the surfactant system.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Beth J. Allison
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Sage G. Ford
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sandra Orgeig
- UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Dino A. Giussani
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
22
|
Domingues RR, Wiltbank MC, Hernandez LL. Maternal serotonin: implications for the use of selective serotonin reuptake inhibitors during gestation†. Biol Reprod 2023; 109:17-28. [PMID: 37098165 PMCID: PMC10344603 DOI: 10.1093/biolre/ioad046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 04/27/2023] Open
Abstract
Maternal use of antidepressants has increased throughout the last decades; selective serotonin reuptake inhibitors (SSRI) are the most prescribed antidepressants. Despite the widespread use of SSRI by women during reproductive age and pregnant women, an increasing amount of research warns of possible detrimental effects of maternal use of SSRI during pregnancy including low birthweight/small for gestational age and preterm birth. In this review, we revisited the impact of maternal use of SSRI during pregnancy, its impact on serotonin homeostasis in the maternal and fetal circulation and the placenta, and its impact on pregnancy outcomes-particularly intrauterine growth restriction and preterm birth. Maternal use of SSRI increases maternal and fetal serotonin. The increase in maternal circulating serotonin and serotonin signaling likely promotes vasoconstriction of the uterine and placental vascular beds decreasing blood perfusion to the uterus and consequently to the placenta and fetus with potential impact on placental function and fetal development. Several adverse pregnancy outcomes are similar between women, sheep, and rodents (decreased placental size, decreased birthweight, shorter gestation length/preterm birth, neonatal morbidity, and mortality) highlighting the importance of animal studies to assess the impacts of SSRI. Herein, we address the complex interactions between maternal SSRI use during gestation, circulating serotonin, and the regulation of blood perfusion to the uterus and fetoplacental unit, fetal growth, and pregnancy complications.
Collapse
Affiliation(s)
- Rafael R Domingues
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Milo C Wiltbank
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Laura L Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
23
|
Sah N, Stenhouse C, Halloran KM, Moses RM, Seo H, Burghardt RC, Johnson GA, Wu G, Bazer FW. Creatine metabolism at the uterine-placental interface throughout gestation in sheep†. Biol Reprod 2023; 109:107-118. [PMID: 37171613 DOI: 10.1093/biolre/ioad052] [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: 11/10/2022] [Revised: 02/21/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023] Open
Abstract
The placenta requires high levels of adenosine triphosphate to maintain a metabolically active state throughout gestation. The creatine-creatine kinase-phosphocreatine system is known to buffer adenosine triphosphate levels; however, the role(s) creatine-creatine kinase-phosphocreatine system plays in uterine and placental metabolism throughout gestation is poorly understood. In this study, Suffolk ewes were ovariohysterectomized on Days 30, 50, 70, 90, 110 and 125 of gestation (n = 3-5 ewes/per day, except n = 2 on Day 50) and uterine and placental tissues subjected to analyses to measure metabolites, mRNAs, and proteins related to the creatine-creatine kinase-phosphocreatine system. Day of gestation affected concentrations and total amounts of guanidinoacetate and creatine in maternal plasma, amniotic fluid and allantoic fluid (P < 0.05). Expression of mRNAs for arginine:glycine amidinotransferase, guanidinoacetate methyltransferase, creatine kinase B, and solute carrier 16A12 in endometria and for arginine:glycine amidinotransferase and creatine kinase B in placentomes changed significantly across days of gestation (P < 0.05). The arginine:glycine amidinotransferase protein was more abundant in uterine luminal epithelium on Days 90 and 125 compared to Days 30 and 50 (P < 0.01). The chorionic epithelium of placentomes expressed guanidinoacetate methyltransferase and solute carrier 6A13 throughout gestation. Creatine transporter (solute carrier 6A8) was expressed by the uterine luminal epithelium and trophectoderm of placentomes throughout gestation. Creatine kinase (creatine kinase B and CKMT1) proteins were localized primarily to the uterine luminal epithelium and to the placental chorionic epithelium of placentomes throughout gestation. Collectively, these results demonstrate cell-specific and temporal regulation of components of the creatine-creatine kinase-phosphocreatine system that likely influence energy homeostasis for fetal-placental development.
Collapse
Affiliation(s)
- Nirvay Sah
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Claire Stenhouse
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | | | - Robyn M Moses
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Heewon Seo
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Gregory A Johnson
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
24
|
Espinoza J. Evolutionary perspective of uteroplacental malperfusion: subjacent insult common to most pregnancy complications. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:7-13. [PMID: 37391928 DOI: 10.1002/uog.26218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 07/02/2023]
Abstract
Linked article: There is a comment on this article by Yagel et al. Click here to view the Correspondence.
Collapse
Affiliation(s)
- J Espinoza
- The Fetal Center, Children's Memorial Hermann Hospital, Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Fetal Intervention, UTHealth, McGovern Medical School, University of Texas, Houston, TX, USA
| |
Collapse
|
25
|
Darby JR, Zhang S, Holman SL, Muhlhausler BS, McMillen IC, Morrison JL. Cardiac growth and metabolism of the fetal sheep are not vulnerable to a 10 day increase in fetal glucose and insulin concentrations during late gestation. Heliyon 2023; 9:e18292. [PMID: 37519661 PMCID: PMC10372399 DOI: 10.1016/j.heliyon.2023.e18292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Aims To evaluate the effects of fetal glucose infusion in late gestation on the mRNA expression and protein abundance of molecules involved in the regulation of cardiac growth and metabolism. Main methods Either saline or glucose was infused into fetal sheep from 130 to 140 days (d) gestation (term, 150 d). At 140 d gestation, left ventricle tissue samples were collected. Quantitative real-time RT-PCR and Western blot were used to determine the mRNA expression and protein abundance of key signalling molecules within the left ventricle of the fetal heart. Key findings Although intra-fetal glucose infusion increased fetal plasma glucose and insulin concentrations, there was no change in the expression of molecules within the signalling pathways that regulate proliferation, hypertrophy, apoptosis or fibrosis in the fetal heart. Cardiac Solute carrier family 2 member 1 (SLC2A1) mRNA expression was decreased by glucose infusion. Glucose infusion increased cardiac mRNA expression of both Peroxisome proliferator activated receptor alpha (PPARA) and peroxisome proliferator activated receptor gamma (PPARG). However, there was no change in the mRNA expression of PPAR cofactors or molecules with PPAR response elements. Furthermore, glucose infusion did not impact the protein abundance of the 5 oxidative phosphorylation complexes of the electron transport chain. Significance Despite a 10-day doubling of fetal plasma glucose and insulin concentrations, the present study suggests that within the fetal left ventricle, the mRNA and protein expression of the signalling molecules involved in cardiac growth, development and metabolism are relatively unaffected.
Collapse
Affiliation(s)
| | | | | | | | | | - Janna L. Morrison
- Corresponding author. Australian Research Council Future Fellow, Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia,
| |
Collapse
|
26
|
Muroya S, Otomaru K, Oshima K, Oshima I, Ojima K, Gotoh T. DNA Methylation of Genes Participating in Hepatic Metabolisms and Function in Fetal Calf Liver Is Altered by Maternal Undernutrition during Gestation. Int J Mol Sci 2023; 24:10682. [PMID: 37445858 DOI: 10.3390/ijms241310682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
This study aimed to elucidate the effects of maternal undernutrition (MUN) on epigenetic modification of hepatic genes in Japanese Black fetal calves during gestation. Using a previously established experimental design feeding the dams with 60% (LN) or 120% (HN) of their global nutritional requirements during the 8.5-month gestational period, DNA methylation in the fetal liver was analyzed with reduced representation bisulfite sequencing (RRBS). The promoters and gene bodies in the LN fetuses were hypomethylated compared to HN fetuses. Pathway analysis showed that the genes with DMR in the exon/intron in the LN group were associated with pathways involved in Cushing syndrome, gastric acid secretion, and aldosterone synthesis and secretion. Promoter hypomethylation in the LN group was frequently observed in genes participating in various signaling pathways (thyroid hormone, Ras/Rap1, PIK3-Akt, cAMP), fatty acid metabolism, and cholesterol metabolism. The promoter hypomethylated genes ALPL and GNAS were upregulated in the LN group, whereas the promoter hypermethylated genes GRB10 and POR were downregulated. The intron/exon hypomethylated genes IGF2, IGF2R, ACAD8, TAT, RARB, PINK1, and SOAT2 were downregulated, whereas the hypermethylated genes IGF2BP2, NOS3, and NR2F1 were upregulated. Collectively, MUN alters the promoter and gene body methylation of genes associated with hepatic metabolisms (energy, cholesterol, mitochondria) and function, suggesting an impact of altered gene methylation on the dysregulation of gene expression in the fetal liver.
Collapse
Affiliation(s)
- Susumu Muroya
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Tsukuba 305-0901, Ibaraki, Japan
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Kazunaga Oshima
- Division of Year-Round Grazing Research, NARO Western Region Agricultural Research Center, 60 Yoshinaga, Ohda 694-0013, Shimane, Japan
| | - Ichiro Oshima
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Koichi Ojima
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Tsukuba 305-0901, Ibaraki, Japan
| | - Takafumi Gotoh
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| |
Collapse
|
27
|
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
|
28
|
Hord TK, Tanner AR, Kennedy VC, Lynch CS, Winger QA, Rozance PJ, Anthony RV. Impact of Chorionic Somatomammotropin In Vivo RNA Interference Phenotype on Uteroplacental Expression of the IGF Axis. Life (Basel) 2023; 13:1261. [PMID: 37374044 PMCID: PMC10302269 DOI: 10.3390/life13061261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
While fetal growth is dependent on many factors, optimal placental function is a prerequisite for a normal pregnancy outcome. The majority of fetal growth-restricted (FGR) pregnancies result from placental insufficiency (PI). The insulin-like growth factors (IGF1 and IGF2) stimulate fetal growth and placental development and function. Previously, we demonstrated that in vivo RNA interference (RNAi) of the placental hormone, chorionic somatomammotropin (CSH), resulted in two phenotypes. One phenotype exhibits significant placental and fetal growth restriction (PI-FGR), impaired placental nutrient transport, and significant reductions in umbilical insulin and IGF1. The other phenotype does not exhibit statistically significant changes in placental or fetal growth (non-FGR). It was our objective to further characterize these two phenotypes by determining the impact of CSH RNAi on the placental (maternal caruncle and fetal cotyledon) expression of the IGF axis. The trophectoderm of hatched blastocysts (9 days of gestation, dGA) were infected with a lentivirus expressing either a non-targeting sequence (NTS RNAi) control or CSH-specific shRNA (CSH RNAi) prior to embryo transfer into synchronized recipient ewes. At ≈125 dGA, pregnancies were fitted with vascular catheters to undergo steady-state metabolic studies. Nutrient uptakes were determined, and tissues were harvested at necropsy. In both CSH RNAi non-FGR and PI-FGR pregnancies, uterine blood flow was significantly reduced (p ≤ 0.05), while umbilical blood flow (p ≤ 0.01), both uterine and umbilical glucose and oxygen uptakes (p ≤ 0.05), and umbilical concentrations of insulin and IGF1 (p ≤ 0.05) were reduced in CSH RNAi PI-FGR pregnancies. Fetal cotyledon IGF1 mRNA concentration was reduced (p ≤ 0.05) in CSH RNAi PI-FGR pregnancies, whereas neither IGF1 nor IGF2 mRNA concentrations were impacted in the maternal caruncles, and either placental tissue in the non-FGR pregnancies. Fetal cotyledon IGF1R and IGF2R mRNA concentrations were not impacted for either phenotype, yet IGF2R was increased (p ≤ 0.01) in the maternal caruncles of CSH RNAi PI-FGR pregnancies. For the IGF binding proteins (IGFBP1, IGFBP2, IGFBP3), only IGFBP2 mRNA concentrations were impacted, with elevated IGFBP2 mRNA in both the fetal cotyledon (p ≤ 0.01) and maternal caruncle (p = 0.08) of CSH RNAi non-FGR pregnancies. These data support the importance of IGF1 in placental growth and function but may also implicate IGFBP2 in salvaging placental growth in non-FGR pregnancies.
Collapse
Affiliation(s)
- Taylor K. Hord
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Amelia R. Tanner
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Victoria C. Kennedy
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Cameron S. Lynch
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Quinton A. Winger
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Paul J. Rozance
- Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Russell V. Anthony
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
29
|
Hu W, Wang X, Bi Y, Bao J, Shang M, Zhang L. The Molecular Mechanism of the TEAD1 Gene and miR-410-5p Affect Embryonic Skeletal Muscle Development: A miRNA-Mediated ceRNA Network Analysis. Cells 2023; 12:cells12060943. [PMID: 36980284 PMCID: PMC10047409 DOI: 10.3390/cells12060943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Muscle development is a complex biological process involving an intricate network of multiple factor interactions. Through the analysis of transcriptome data and molecular biology confirmation, this study aims to reveal the molecular mechanism underlying sheep embryonic skeletal muscle development. The RNA sequencing of embryos was conducted, and microRNA (miRNA)-mediated competitive endogenous RNA (ceRNA) networks were constructed. qRT-PCR, siRNA knockdown, CCK-8 assay, scratch assay, and dual luciferase assay were used to carry out gene function identification. Through the analysis of the ceRNA networks, three miRNAs (miR-493-3p, miR-3959-3p, and miR-410-5p) and three genes (TEAD1, ZBTB34, and POGLUT1) were identified. The qRT-PCR of the DE-miRNAs and genes in the muscle tissues of sheep showed that the expression levels of the TEAD1 gene and miR-410-5p were correlated with the growth rate. The knockdown of the TEAD1 gene by siRNA could significantly inhibit the proliferation of sheep primary embryonic myoblasts, and the expression levels of SLC1A5, FoxO3, MyoD, and Pax7 were significantly downregulated. The targeting relationship between miR-410-5p and the TEAD1 gene was validated by a dual luciferase assay, and miR-410-5p can significantly downregulate the expression of TEAD1 in sheep primary embryonic myoblasts. We proved the regulatory relationship between miR-410-5p and the TEAD1 gene, which was related to the proliferation of sheep embryonic myoblasts. The results provide a reference and molecular basis for understanding the molecular mechanism of embryonic muscle development.
Collapse
Affiliation(s)
- Wenping Hu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xinyue Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yazhen Bi
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jingjing Bao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Mingyu Shang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Li Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
30
|
Tajonar K, Gonzalez-Ronquillo M, Relling A, Nordquist RE, Nawroth C, Vargas-Bello-Pérez E. Toward assessing the role of dietary fatty acids in lamb's neurological and cognitive development. Front Vet Sci 2023; 10:1081141. [PMID: 36865439 PMCID: PMC9971820 DOI: 10.3389/fvets.2023.1081141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 02/16/2023] Open
Abstract
Understanding and measuring sheep cognition and behavior can provide us with measures to safeguard the welfare of these animals in production systems. Optimal neurological and cognitive development of lambs is important to equip individuals with the ability to better cope with environmental stressors. However, this development can be affected by nutrition with a special role from long-chain fatty acid supply from the dam to the fetus or in lamb's early life. Neurological development in lambs takes place primarily during the first two trimesters of gestation. Through late fetal and early postnatal life, the lamb brain has a high level of cholesterol synthesis. This rate declines rapidly at weaning and remains low throughout adulthood. The main polyunsaturated fatty acids (PUFA) in the brain are ω-6 arachidonic acid and ω-3 docosahexaenoic acid (DHA), which are elements of plasma membranes' phospholipids in neuronal cells. DHA is essential for keeping membrane integrity and is vital for normal development of the central nervous system (CNS), and its insufficiency can damage cerebral functions and the development of cognitive capacities. In sheep, there is evidence that supplying PUFA during gestation or after birth may be beneficial to lamb productive performance and expression of species-specific behaviors. The objective of this perspective is to discuss concepts of ruminant behavior and nutrition and reflect on future research directions that could help to improve our knowledge on how dietary fatty acids (FA) relate to optimal neurological and cognitive development in sheep.
Collapse
Affiliation(s)
- Karen Tajonar
- Departamento de Medicina y Zootecnia de Rumiantes, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico,Department of Animal Sciences, School of Agriculture, Policy and Development, University of Reading, Reading, United Kingdom
| | - Manuel Gonzalez-Ronquillo
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Alejandro Relling
- Department of Animal Science, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, United States
| | - Rebecca E. Nordquist
- Unit Animals in Science and Society, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Christian Nawroth
- Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany,*Correspondence: Christian Nawroth ✉
| | - Einar Vargas-Bello-Pérez
- Department of Animal Sciences, School of Agriculture, Policy and Development, University of Reading, Reading, United Kingdom,Einar Vargas-Bello-Pérez ✉
| |
Collapse
|
31
|
Meakin AS, Amirmostofian M, Darby JRT, Holman SL, Morrison JL, Wiese MD. Characterisation of cytochrome P450 isoenzyme activity in sheep liver and placental microsomes. Placenta 2023; 131:82-89. [PMID: 36527743 DOI: 10.1016/j.placenta.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Drug metabolism during pregnancy is a complex process that involves maternal, placental and fetal sites of metabolism. Indeed, there is a lack of clarity provided from drug metabolism in human pregnancy due to ethical limitations. Large animal models of human pregnancy provide an opportunity to quantify activity of phase 1 drug metabolism mediated by cytochrome P450 (CYP) enzymes in the maternal, placental, and fetal compartments. Herein, we have validated a comprehensive assay to quantify maternal, placental, and fetal CYP activity. METHODS Isolated microsomes from sheep maternal liver, placenta, and fetal liver (140d gestation, term = 150d) were incubated with CYP-specific probe drugs to quantify the activity of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A. Inhibition studies were performed to validate specificity of probe drugs. The validated assay was developed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS CYP1A2, CYP2B6, CYP2C8, CYP2C19, CYP2D6, CYP2E1 and CYP3A were active in maternal liver. In contrast, only CYP1A2, CYP2C8 and CYP2D6 were active in the placenta, whereas CYP2B6, CYP2C8 and CYP2D6 were active in the fetal liver. Of the placental-specific CYPs validated, CYP1A2 increased in type A compared with type D placentomes, whereas CYP2C8 activity increased in type B compared with type A and C. DISCUSSION This study has established conditions for compartment-specific CYP activity in the sheep maternal-placental-fetal unit using a validated and standardised experimental workflow. Compartment- and placentome type-specific CYP activity are important considerations when examining drug metabolism in the maternal-placental-fetal unit and in determining the impact of pregnancy complications.
Collapse
Affiliation(s)
- 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
| | - Marzieh Amirmostofian
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack RT 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
| | - 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
| | - 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.
| | - Michael D Wiese
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
32
|
Deluao JC, Winstanley Y, Robker RL, Pacella-Ince L, Gonzalez MB, McPherson NO. OXIDATIVE STRESS AND REPRODUCTIVE FUNCTION: Reactive oxygen species in the mammalian pre-implantation embryo. Reproduction 2022; 164:F95-F108. [PMID: 36111646 DOI: 10.1530/rep-22-0121] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
In brief Reactive oxygen species are generated throughout the pre-implantation period and are necessary for normal embryo formation. However, at pathological levels, they result in reduced embryo viability which can be mediated through factors delivered by sperm and eggs at conception or from the external environment. Abstract Reactive oxygen species (ROS) occur naturally in pre-implantation embryos as a by-product of ATP generation through oxidative phosphorylation and enzymes such as NADPH oxidase and xanthine oxidase. Biological concentrations of ROS are required for crucial embryonic events such as pronuclear formation, first cleavage and cell proliferation. However, high concentrations of ROS are detrimental to embryo development, resulting in embryo arrest, increased DNA damage and modification of gene expression leading to aberrant fetal growth and health. In vivo embryos are protected against oxidative stress by oxygen scavengers present in follicular and oviductal fluids, while in vitro, embryos rely on their own antioxidant defence mechanisms to protect against oxidative damage, including superoxide dismutase, catalase, glutathione and glutamylcysteine synthestase. Pre-implantation embryonic ROS originate from eggs, sperm and embryos themselves or from the external environment (i.e. in vitro culture system, obesity and ageing). This review examines the biological and pathological roles of ROS in the pre-implantation embryo, maternal and paternal origins of embryonic ROS, and from a clinical perspective, we comment on the growing interest in combating increased oxidative damage in the pre-implantation embryo through the addition of antioxidants.
Collapse
Affiliation(s)
- Joshua C Deluao
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Yasmyn Winstanley
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Rebecca L Robker
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Leanne Pacella-Ince
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Repromed, Dulwich, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Nicole O McPherson
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Repromed, Dulwich, Australia
| |
Collapse
|
33
|
Altered Cord Blood Lipid Concentrations Correlate with Birth Weight and Doppler Velocimetry of Fetal Vessels in Human Fetal Growth Restriction Pregnancies. Cells 2022; 11:cells11193110. [PMID: 36231072 PMCID: PMC9562243 DOI: 10.3390/cells11193110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Fetal growth restriction (FGR) is associated with short- and long-term morbidity, often with fetal compromise in utero, evidenced by abnormal Doppler velocimetry of fetal vessels. Lipids are vital for growth and development, but metabolism in FGR pregnancy, where fetuses do not grow to full genetic potential, is poorly understood. We hypothesize that triglyceride concentrations are increased in placentas and that important complex lipids are reduced in cord plasma from pregnancies producing the smallest babies (birth weight < 5%) and correlate with ultrasound Dopplers. Dopplers (umbilical artery, UA; middle cerebral artery, MCA) were assessed longitudinally in pregnancies diagnosed with estimated fetal weight (EFW) < 10% at ≥29 weeks gestation. For a subset of enrolled women, placentas and cord blood were collected at delivery, fatty acids were extracted and targeted lipid class analysis (triglyceride, TG; phosphatidylcholine, PC; lysophosphatidylcholine, LPC; eicosanoid) performed by LCMS. For this sub-analysis, participants were categorized as FGR (Fenton birth weight, BW ≤ 5%) or SGA "controls" (Fenton BW > 5%). FGRs (n = 8) delivered 1 week earlier (p = 0.04), were 29% smaller (p = 0.002), and had 133% higher UA pulsatility index (PI, p = 0.02) than SGAs (n = 12). FGR plasma TG, free arachidonic acid (AA), and several eicosanoids were increased (p < 0.05); docosahexaenoic acid (DHA)-LPC was decreased (p < 0.01). Plasma TG correlated inversely with BW (p < 0.05). Plasma EET, non-esterified AA, and DHA correlated inversely with BW and directly with UA PI (p < 0.05). Placental DHA-PC and AA-PC correlated directly with MCA PI (p < 0.05). In fetuses initially referred for inadequate fetal growth (EFW < 10%), those with BW ≤ 5% demonstrated distinctly different cord plasma lipid profiles than those with BW > 5%, which correlated with Doppler PIs. This provides new insights into fetal lipidomic response to the FGR in utero environment. The impact of these changes on specific processes of growth and development (particularly fetal brain) have not been elucidated, but the relationship with Doppler PI may provide additional context for FGR surveillance, and a more targeted approach to nutritional management of these infants.
Collapse
|
34
|
Flouri D, Darby JRT, Holman SL, Cho SKS, Dimasi CG, Perumal SR, Ourselin S, Aughwane R, Mufti N, Macgowan CK, Seed M, David AL, Melbourne A, Morrison JL. Placental MRI Predicts Fetal Oxygenation and Growth Rates in Sheep and Human Pregnancy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203738. [PMID: 36031385 PMCID: PMC9596844 DOI: 10.1002/advs.202203738] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Indexed: 06/09/2023]
Abstract
Magnetic resonance imaging (MRI) assessment of fetal blood oxygen saturation (SO2 ) can transform the clinical management of high-risk pregnancies affected by fetal growth restriction (FGR). Here, a novel MRI method assesses the feasibility of identifying normally grown and FGR fetuses in sheep and is then applied to humans. MRI scans are performed in pregnant ewes at 110 and 140 days (term = 150d) gestation and in pregnant women at 28+3 ± 2+5 weeks to measure feto-placental SO2 . Birth weight is collected and, in sheep, fetal blood SO2 is measured with a blood gas analyzer (BGA). Fetal arterial SO2 measured by BGA predicts fetal birth weight in sheep and distinguishes between fetuses that are normally grown, small for gestational age, and FGR. MRI feto-placental SO2 in late gestation is related to fetal blood SO2 measured by BGA and body weight. In sheep, MRI feto-placental SO2 in mid-gestation is related to fetal SO2 later in gestation. MRI feto-placental SO2 distinguishes between normally grown and FGR fetuses, as well as distinguishing FGR fetuses with and without normal Doppler in humans. Thus, a multi-compartment placental MRI model detects low placental SO2 and distinguishes between small hypoxemic fetuses and normally grown fetuses.
Collapse
Affiliation(s)
- Dimitra Flouri
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonSE1 7EUUK
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonWC1E 6BTUK
| | - Jack R. T. Darby
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Stacey L. Holman
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Steven K. S. Cho
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
- Department of PhysiologyThe Hospital for Sick ChildrenUniversity of TorontoTorontoON M5G 1X8Canada
| | - Catherine G. Dimasi
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Sunthara R. Perumal
- South Australian Health & Medical Research InstitutePreclinicalImaging & Research LaboratoriesAdelaideSA 5001Australia
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonSE1 7EUUK
| | - Rosalind Aughwane
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonWC1E 6BTUK
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonWC1E 6AUUK
| | - Nada Mufti
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonWC1E 6BTUK
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonWC1E 6AUUK
| | - Christopher K. Macgowan
- Division of Translational MedicineThe Hospital for Sick ChildrenUniversity of TorontoTorontoON M5G 1X8Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoON M5S 1A1Canada
| | - Mike Seed
- Department of PaediatricsDivision of CardiologyThe Hospital for Sick ChildrenUniversity of TorontoTorontoON M5G 1X8Canada
- Department of Diagnostic ImagingThe Hospital for Sick ChildrenUniversity of TorontoTorontoON M5G 1X8Canada
| | - Anna L. David
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonWC1E 6AUUK
- NIHR Biomedical Research CentreUniversity College London HospitalsLondonW1T 7DNUK
| | - Andrew Melbourne
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonSE1 7EUUK
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonWC1E 6BTUK
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Janna L. Morrison
- Early Origins of Adult Health Research GroupHealth and Biomedical InnovationUniSA Clinical and Health SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| |
Collapse
|
35
|
Domingues RR, Beard AD, Connelly MK, Wiltbank MC, Hernandez LL. Fluoxetine-induced perinatal morbidity in a sheep model. Front Med (Lausanne) 2022; 9:955560. [PMID: 35991651 PMCID: PMC9386076 DOI: 10.3389/fmed.2022.955560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRI) are the most common antidepressants used by pregnant women. However, adverse pregnancy outcomes have been described in women taking SSRI during pregnancy—placental lesions, premature birth, poor neonatal adaptation. We aimed to investigate the effects of fluoxetine (Prozac® most commonly used SSRI) treatment during the last month of gestation on pregnancy complications, placental and neonatal health in a non-depressed sheep model. On day 119 ± 1 postbreeding (experimental day 0; E0) of a 151-day expected gestation, Hampshire ewes were randomly assigned to receive fluoxetine (n = 9 ewes, 15 lambs; daily intravenously treatment with 10 mg/kg on E0 and E1 and 5 mg/kg daily thereafter until parturition) or to a control group (n = 10; 14 lambs; vehicle only). Blood samples from ewes were collected throughout the experimental period and postpartum; blood from lambs were collected postpartum. Analysis of variance was used for statistical analysis. Fluoxetine treatment reduced placentome growth during the last month of pregnancy. Gestation length was decreased by 4.5 days in fluoxetine-treated ewes. Birthweight was reduced in lambs exposed to fluoxetine in utero; weights remained decreased until postnatal day 3. Placentome diameter by birthweight ratio was not different between groups suggesting that the decreased placentome diameter was accompanied by decreased lamb birthweight. During the first week postnatal, lambs exposed to fluoxetine in utero had decreased blood pH and decreased total carbon dioxide, bicarbonate, and base excess and increased lactate (days 3–6), collectively indicative of metabolic acidemia. Additionally, ionized calcium was decreased between postnatal days 0 to 4 in lambs exposed to fluoxetine in utero. Using a non-depressed animal model clearly defines a role for SSRI on the occurrence of perinatal complications and neonatal morbidity. The decreased placentome diameter, shortened gestation, decreased birthweight, decreased calcium levels, and neonatal acidemia suggest the occurrence of intrauterine growth restriction. The persistence of neonatal acidemia for several days postpartum suggests poor neonatal adaptation to extrauterine environment.
Collapse
Affiliation(s)
- Rafael R. Domingues
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Adam D. Beard
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Meghan K. Connelly
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Milo C. Wiltbank
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Laura L. Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
- *Correspondence: Laura L. Hernandez
| |
Collapse
|
36
|
Valenzuela I, Kinoshita M, van der Merwe J, Maršál K, Deprest J. Prenatal interventions for fetal growth restriction in animal models: A systematic review. Placenta 2022; 126:90-113. [PMID: 35796064 DOI: 10.1016/j.placenta.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/20/2022] [Accepted: 06/17/2022] [Indexed: 12/09/2022]
Abstract
Fetal growth restriction (FGR) in human pregnancy is associated with perinatal mortality, short- and long-term morbidities. No prenatal therapy is currently established despite decades of research. We aimed to review interventions in animal models for prenatal FGR treatment, and to seek the next steps for an effective clinical therapy. We registered our protocol and searched MEDLINE, Embase, and The Cochrane Library with no language restrictions, in accordance with the PRISMA guideline. We included all studies that reported the effects of any prenatal intervention in animal models of induced FGR. From 3257 screened studies, 202 describing 237 interventions were included for the final synthesis. Mice and rats were the most used animals (79%) followed by sheep (16%). Antioxidants (23%), followed by vasodilators (18%), nutrients (14%), and immunomodulators (12%) were the most tested therapy. Two-thirds of studies only reported delivery or immediate neonatal outcomes. Adverse effects were rarely reported (11%). Most studies (73%), independent of the intervention, showed a benefit in fetal survival or birthweight. The risk of bias was high, mostly due to the lack of randomization, allocation concealment, and blinding. Future research should aim to describe both short- and long-term outcomes across various organ systems in well-characterized models. Further efforts must be made to reduce selection, performance, and detection bias.
Collapse
|
37
|
Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022; 125:4-9. [PMID: 35414477 PMCID: PMC10947607 DOI: 10.1016/j.placenta.2022.03.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 01/16/2023]
Abstract
Pregnancy complications adversely impact both mother and/or fetus throughout the lifespan. Fetal growth restriction (FGR) occurs when a fetus fails to reach their intrauterine potential for growth, it is the second highest leading cause of infant mortality, and leads to increased risk of developing non-communicable diseases in later life due 'fetal programming'. Abnormal placental development, growth and/or function underlies approximately 75% of FGR cases and there is currently no treatment save delivery, often prematurely. We previously demonstrated in a murine model of FGR that nanoparticle mediated, intra-placental human IGF-1 gene therapy maintains normal fetal growth. Multiple models of FGR currently exist reflecting the etiologies of human FGR and have been used by us and others to investigate the development of in utero therapeutics as discussed here. In addition to the in vivo models discussed herein, utilizing human models including in vitro (Choriocarcinoma cell lines and primary trophoblasts) and ex vivo (term villous fragments and placenta cotyledon perfusion) we have demonstrated robust nanoparticle uptake, transgene expression, nutrient transporter regulation without transfer to the fetus. For translational gene therapy application in the human placenta, there are multiple avenues that require investigation including syncytial uptake from the maternal circulation, transgene expression, functionality and longevity of treatment, impact of treatment on the mother and developing fetus. The potential impact of treating the placenta during gestation is high, wide-ranging across pregnancy complications, and may offer reduced risk of developing associated cardio-metabolic diseases in later life impacting at both an individual and societal level.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States.
| |
Collapse
|
38
|
Meakin AS, Darby JR, Holman SL, Wiese MD, Morrison JL. Maternal-placental-fetal drug metabolism is altered by late gestation undernutrition in the pregnant ewe. Life Sci 2022; 298:120521. [DOI: 10.1016/j.lfs.2022.120521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
|
39
|
Sheep as a model for neuroendocrinology research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:1-34. [PMID: 35595346 DOI: 10.1016/bs.pmbts.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.
Collapse
|
40
|
Vautier AN, Cadaret CN. Long-Term Consequences of Adaptive Fetal Programming in Ruminant Livestock. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.778440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Environmental perturbations during gestation can alter fetal development and postnatal animal performance. In humans, intrauterine growth restriction (IUGR) resulting from adaptive fetal programming is known as a leading cause of perinatal morbidity and mortality and predisposes offspring to metabolic disease, however, the prevalence and impact in livestock is not characterized as well. Multiple animal models have been developed as a proxy to determine mechanistic changes that underlie the postnatal phenotype resulting from these programming events in humans but have not been utilized as robustly in livestock. While the overall consequences are similar between models, the severity of the conditions appear to be dependent on type, timing, and duration of insult, indicating that some environmental insults are of more relevance to livestock production than others. Thus far, maternofetal stress during gestation has been shown to cause increased death loss, low birth weight, inefficient growth, and aberrant metabolism. A breadth of this data comes from the fetal ruminant collected near term or shortly thereafter, with fewer studies following these animals past weaning. Consequently, even less is known about how adaptive fetal programming impacts subsequent progeny. In this review, we summarize the current knowledge of the postnatal phenotype of livestock resulting from different models of fetal programming, with a focus on growth, metabolism, and reproductive efficiency. We further describe what is currently known about generational impacts of fetal programming in production systems, along with gaps and future directions to consider.
Collapse
|
41
|
Chand KK, Pannek K, Colditz PB, Wixey JA. Brain outcomes in runted piglets: a translational model of fetal growth restriction. Dev Neurosci 2022; 44:194-204. [PMID: 35263744 DOI: 10.1159/000523995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/07/2022] [Indexed: 11/19/2022] Open
Abstract
etal growth restriction (FGR) is associated with long-term neurodevelopmental disabilities including learning and behavioural disorders, autism, and cerebral palsy. Persistent changes in brain structure and function that are associated with developmental disabilities are demonstrated in FGR neonates. However, the mechanisms underlying these changes remain to be determined. There are currently no therapeutic interventions available to protect the FGR newborn brain. With the wide range of long-term neurodevelopmental disorders associated with FGR, the use of an animal model appropriate to investigating mechanisms of injury in the FGR newborn is crucial for the development of effective and targeted therapies for babies. Piglets are ideal animals to explore how perinatal insults affect brain structure and function. FGR occurs spontaneously in the piglet, unlike other animal models that require surgical or chemical intervention, allowing brain outcomes to be studied without the confounding impacts of experimental interventions. The FGR piglet mimics many of the human pathophysiological outcomes associated with FGR including asymmetrical growth restriction with brain sparing. This review will discuss the similarities observed in brain outcomes between the human FGR and FGR piglet from a magnetic resonance imaging in the living and a histological perspective. FGR piglet studies provide the opportunity to determine and track mechanisms of brain injury in a clinically relevant animal model of FGR. Findings from these FGR piglet studies may provide critical information to rapidly translate neuroprotective interventions to clinic to improve outcomes for newborn babies.
Collapse
Affiliation(s)
- Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Kerstin Pannek
- The Australian E-Health Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Perinatal Research Centre, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
42
|
Christiansen F, Uhart MM, Bejder L, Clapham P, Ivashchenko Y, Tormosov D, Lewin N, Sironi M. Foetal growth, birth size and energetic cost of gestation in southern right whales. J Physiol 2022; 600:2245-2266. [PMID: 35261040 PMCID: PMC9322553 DOI: 10.1113/jp282351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/02/2022] [Indexed: 11/08/2022] Open
Abstract
Abstract The cost of reproduction greatly affects a species’ life history strategy. Baleen whales exhibit some of the fastest offspring growth rates in the animal kingdom. We quantified the energetic cost of gestation for southern right whales (Eubalaena australis) by combining whaling catch records of pregnant females with photogrammetry data on southern right whale mothers and calves from two breeding grounds in Argentina and Australia. The relationship between calf birth size and maternal length was determined from repeated measurements of individual females before and after giving birth. Fetal growth was determined from generalized linear models fitted to fetal length data from whaling operations between 1961 and 1967. Fetal length was converted to volume and mass, using the volume‐to‐length relationship of newborn southern right whales calves, and published tissue composition and energy content estimates. Fetal maintenance costs (heat of gestation) and the energy content of the placenta were predicted from published relationships and added to the fetal growth cost to calculate the total cost of gestation. Our findings showed that fetal growth rates and birth size increased linearly with maternal length, with calves being born at ∼35% maternal length. Fetal length increased curvilinearly through gestation, which resulted in an exponential increase in fetal volume and mass. Consequently, the cost of gestation was very low during the first (0.1% of total cost) and second trimester (4.9%), but increased rapidly during the last trimester (95.0%). The heat of gestation incurred the highest cost for pregnant females (73.8%), followed by fetal growth (21.2%) and the placental energy content (5.0%). Key points Baleen whales exhibit some of the fastest fetal growth rates in the animal kingdom. Despite this, the energetic cost of gestation is largely unknown, as well as the influence of maternal body size on fetal growth rates and calf birth sizes. We combined historical whaling records and drone photogrammetry data to determine fetal growth rates and birth sizes in southern right whales (Eubalaena australis), from which we estimated the cost of gestation. Calf birth size, and consequent fetal growth rates, increased positively with maternal body size. The cost of gestation was negligible for southern right whale females during the first two trimesters, but increased rapidly during the last trimester. These results show that late gestation incurs a significant cost for baleen whale females, and needs to be accounted for in bioenergetic models.
Collapse
Affiliation(s)
- Fredrik Christiansen
- Aarhus Institute of Advanced Studies, Høegh-Guldbergs Gade 6B, Aarhus C, 8000, Denmark.,Zoophysiology, Department of Biology, Aarhus University, C.F. Møllers Allé 3, Aarhus C, 8000, Denmark
| | - Marcela M Uhart
- Southern Right Whale Health Monitoring Program, Puerto Madryn, Chubut, 9120, Argentina.,Karen C. Drayer Wildlife Health Center, School of Veterinary Medicine, University of California Davis, Davis, California, 95616, USA
| | - Lars Bejder
- Zoophysiology, Department of Biology, Aarhus University, C.F. Møllers Allé 3, Aarhus C, 8000, Denmark.,Marine Mammal Research Program, Hawaii Institute of Marine Biology, University of Hawaii at Manoa, Kaneohe, HI, 96744, USA
| | | | | | - Dmitry Tormosov
- Seastar Scientific Russia, Kaliningrad, Karl Marx St 76, 236022, Russia
| | - Nicolás Lewin
- Instituto de Conservación de Ballenas, Buenos Aires, Argentina
| | - Mariano Sironi
- Southern Right Whale Health Monitoring Program, Puerto Madryn, Chubut, 9120, Argentina.,Instituto de Conservación de Ballenas, Buenos Aires, Argentina.,Diversidad Animal II, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| |
Collapse
|
43
|
Stremming J, Chang EI, Knaub LA, Armstrong ML, Baker PR, Wesolowski SR, Reisdorph N, Reusch JEB, Brown LD. Lower citrate synthase activity, mitochondrial complex expression, and fewer oxidative myofibers characterize skeletal muscle from growth-restricted fetal sheep. Am J Physiol Regul Integr Comp Physiol 2022; 322:R228-R240. [PMID: 34907787 PMCID: PMC8858669 DOI: 10.1152/ajpregu.00222.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022]
Abstract
Skeletal muscle from the late gestation sheep fetus with intrauterine growth restriction (IUGR) has evidence of reduced oxidative metabolism. Using a sheep model of placental insufficiency and IUGR, we tested the hypothesis that by late gestation, IUGR fetal skeletal muscle has reduced capacity for oxidative phosphorylation because of intrinsic deficits in mitochondrial respiration. We measured mitochondrial respiration in permeabilized muscle fibers from biceps femoris (BF) and soleus (SOL) from control and IUGR fetal sheep. Using muscles including BF, SOL, tibialis anterior (TA), and flexor digitorum superficialis (FDS), we measured citrate synthase (CS) activity, mitochondrial complex subunit abundance, fiber type distribution, and gene expression of regulators of mitochondrial biosynthesis. Ex vivo mitochondrial respiration was similar in control and IUGR muscle. However, CS activity was lower in IUGR BF and TA, indicating lower mitochondrial content, and protein expression of individual mitochondrial complex subunits was lower in IUGR TA and BF in a muscle-specific pattern. IUGR TA, BF, and FDS also had lower expression of type I oxidative fibers. Fiber-type shifts that support glycolytic instead of oxidative metabolism may be advantageous for the IUGR fetus in a hypoxic and nutrient-deficient environment, whereas these adaptions may be maladaptive in postnatal life.
Collapse
Affiliation(s)
- Jane Stremming
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | - Eileen I Chang
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | - Leslie A Knaub
- Division of Endocrinology, University of Colorado, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | | | - Peter R Baker
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | | | | | - Jane E B Reusch
- Division of Endocrinology, University of Colorado, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Laura D Brown
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
44
|
Muroya S, Zhang Y, Otomaru K, Oshima K, Oshima I, Sano M, Roh S, Ojima K, Gotoh T. Maternal Nutrient Restriction Disrupts Gene Expression and Metabolites Associated with Urea Cycle, Steroid Synthesis, Glucose Homeostasis, and Glucuronidation in Fetal Calf Liver. Metabolites 2022; 12:metabo12030203. [PMID: 35323646 PMCID: PMC8949217 DOI: 10.3390/metabo12030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/23/2022] Open
Abstract
This study aimed to understand the mechanisms underlying the effects of maternal undernutrition (MUN) on liver growth and metabolism in Japanese Black fetal calves (8.5 months in utero) using an approach that integrates metabolomics and transcriptomics. Dams were fed 60% (low-nutrition; LN) or 120% (high-nutrition; HN) of their overall nutritional requirements during gestation. We found that MUN markedly decreased the body and liver weights of the fetuses; metabolomic analysis revealed that aspartate, glycerol, alanine, gluconate 6-phosphate, and ophthalmate levels were decreased, whereas UDP-glucose, UDP-glucuronate, octanoate, and 2-hydroxybutyrate levels were decreased in the LN fetal liver (p ≤ 0.05). According to metabolite set enrichment analysis, the highly different metabolites were associated with metabolisms including the arginine and proline metabolism, nucleotide and sugar metabolism, propanoate metabolism, glutamate metabolism, porphyrin metabolism, and urea cycle. Transcriptomic and qPCR analyses revealed that MUN upregulated QRFPR and downregulated genes associated with the glucose homeostasis (G6PC, PCK1, DPP4), ketogenesis (HMGCS2), glucuronidation (UGT1A1, UGT1A6, UGT2A1), lipid metabolism (ANGPTL4, APOA5, FADS2), cholesterol and steroid homeostasis (FDPS, HSD11B1, HSD17B6), and urea cycle (CPS1, ASS1, ASL, ARG2). These metabolic pathways were extracted as relevant terms in subsequent gene ontology/pathway analyses. Collectively, these results indicate that the citrate cycle was maintained at the expense of activities of the energy metabolism, glucuronidation, steroid hormone homeostasis, and urea cycle in the liver of MUN fetuses.
Collapse
Affiliation(s)
- Susumu Muroya
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Ibaraki, Tsukuba 305-0901, Japan;
- Correspondence: (S.M.); (T.G.)
| | - Yi Zhang
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Japan; (Y.Z.); (I.O.)
| | - Kounosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Japan;
| | - Kazunaga Oshima
- Division of Year-Round Grazing Research, NARO Western Region Agricultural Research Center, 60 Yoshinaga, Ohda 694-0013, Shimane, Japan;
| | - Ichiro Oshima
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Japan; (Y.Z.); (I.O.)
| | - Mitsue Sano
- Faculty of Human Culture, University of Shiga Prefecture, 2500 Hassaka-cho, Hikone 522-8533, Shiga, Japan;
| | - Sanggun Roh
- Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Miyagi, Japan;
| | - Koichi Ojima
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Ibaraki, Tsukuba 305-0901, Japan;
| | - Takafumi Gotoh
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Japan; (Y.Z.); (I.O.)
- Correspondence: (S.M.); (T.G.)
| |
Collapse
|
45
|
Drake RR, Louey S, Thornburg KL. Intrauterine growth restriction elevates circulating acylcarnitines and suppresses fatty acid metabolism genes in the fetal sheep heart. J Physiol 2022; 600:655-670. [PMID: 34802149 PMCID: PMC9075772 DOI: 10.1113/jp281415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023] Open
Abstract
At birth, the mammalian myocardium switches from using carbohydrates as the primary energy substrate to free fatty acids as the primary fuel. Thus, a compromised switch could jeopardize normal heart function in the neonate. Placental embolization in sheep is a reliable model of intrauterine growth restriction (IUGR). It leads to suppression of both proliferation and terminal differentiation of cardiomyocytes. We hypothesized that the expression of genes regulating cardiac fatty acid metabolism would be similarly suppressed in IUGR, leading to compromised processing of lipids. Following 10 days of umbilicoplacental embolization in fetal sheep, IUGR fetuses had elevated circulating long-chain fatty acylcarnitines compared with controls (C14: CTRL 0.012 ± 0.005 nmol/ml vs. IUGR 0.018 ± 0.005 nmol/ml, P < 0.05; C18: CTRL 0.027 ± 0.009 nmol/mol vs. IUGR 0.043 ± 0.024 nmol/mol, P < 0.05, n = 12 control, n = 12 IUGR) indicative of impaired fatty acid metabolism. Uptake studies using fluorescently tagged BODIPY-C12-saturated free fatty acid in live, isolated cardiomyocytes showed lipid droplet area and number were not different between control and IUGR cells. mRNA levels of sarcolemmal fatty acid transporters (CD36, FATP6), acylation enzymes (ACSL1, ACSL3), mitochondrial transporter (CPT1), β-oxidation enzymes (LCAD, HADH, ACAT1), tricarboxylic acid cycle enzyme (IDH), esterification enzymes (PAP, DGAT) and regulator of the lipid droplet formation (BSCL2) gene were all suppressed in IUGR myocardium (P < 0.05). However, protein levels for these regulatory genes were not different between groups. This discordance between mRNA and protein levels in the stressed myocardium suggests an adaptive protection of key myocardial enzymes under conditions of placental insufficiency. KEY POINTS: The fetal heart relies on carbohydrates in utero and must be prepared to metabolize fatty acids after birth but the effects of compromised fetal growth on the maturation of this metabolic system are unknown. Plasma fatty acylcarnitines are elevated in intrauterine growth-restricted (IUGR) fetuses compared with control fetuses, indicative of impaired fatty acid metabolism in fetal organs. Fatty acid uptake and storage are not different in IUGR cardiomyocytes compared with controls. mRNA levels of genes regulating fatty acid transporter and metabolic enzymes are suppressed in the IUGR myocardium compared with controls, while protein levels remain unchanged. Mismatches in gene and protein expression, and increased circulating fatty acylcarnitines may have long-term implications for offspring heart metabolism and adult health in IUGR individuals. This requires further investigation.
Collapse
Affiliation(s)
- Rachel R Drake
- Center for Developmental Health, Knight Cardiovascular Institute, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Samantha Louey
- Center for Developmental Health, Knight Cardiovascular Institute, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Kent L Thornburg
- Center for Developmental Health, Knight Cardiovascular Institute, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
46
|
Liu YC, Ma C, Zi Y, He S, Yang H, Zhang M, Gao F. Effects of intrauterine growth restriction during late pregnancy on the cell growth, proliferation, and differentiation in ovine fetal thymuses. Anim Biosci 2022; 35:989-998. [PMID: 35073662 PMCID: PMC9271382 DOI: 10.5713/ab.21.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022] Open
Abstract
Objective This study investigated the effects of intrauterine growth restriction (IUGR) during late pregnancy on the cell growth, proliferation, and differentiation in ovine fetal thymuses. Methods Eighteen time-mated Mongolian ewes with singleton fetuses were allocated to three groups at d 90 of pregnancy: restricted group 1 (RG1, 0.18 MJ ME/body weight [BW]0.75/d, n = 6), restricted group 2 (RG2, 0.33 MJ ME/BW0.75/d, n = 6) and control group (CG, ad libitum, 0.67 MJ ME/BW0.75/d, n = 6). Fetuses were recovered at slaughter on d 140. Results The G0/G1 phase cell number in fetal thymus of the RG1 group was increased but the proliferation index and the expression of proliferating cell nuclear antigen (PCNA) were reduced compared with the CG group (p<0.05). Fetuses in the RG1 group exhibited decreased growth hormone receptor (GHR), insulin-like growth factor 2 receptor (IGF-2R), and their mRNA expressions (p<0.05). For the RG2 fetuses, there were no differences in the proliferation index and PCNA expression (p>0.05), but growth hormone (GH) and the mRNA expression of GHR were lower than those of the CG group (p<0.05). The thymic mRNA expressions of cyclin-dependent protein kinases (CDKs including CDK1, CDK2, and CDK4), CCNE, E2-factors (E2F1, E2F2, and E2F5) were reduced in the RG1 and RG2 groups (p<0.05), and decreased mRNA expressions of E2F4, CCNA, CCNB, and CCND were occurred in the RG1 fetuses (p<0.05). The decreased E-cadherin (E-cad) as a marker for epithelial-mesenchymal transition (EMT) was found in the RG1 and RG2 groups (p< 0.05), but the OB-cadherin which is a marker for activated fibroblasts was increased in fetal thymus of the RG1 group (p<0.05). Conclusion These results indicate that weakened GH/IGF signaling system repressed the cell cycle progression in G0/G1 phase in IUGR fetal thymus, but the switch from reduced E-cad to increased OB-cadherin suggests that transdifferentiation process of EMT associated with fibrogenesis was strengthened. The impaired cell growth, retarded proliferation and modified differentiation were responsible for impaired maturation of IUGR fetal thymus.
Collapse
|
47
|
Mutamba AK, He X, Wang T. Therapeutic advances in overcoming intrauterine growth restriction induced metabolic syndrome. Front Pediatr 2022; 10:1040742. [PMID: 36714657 PMCID: PMC9875160 DOI: 10.3389/fped.2022.1040742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Intrauterine growth restriction (IUGR) remains a great public health challenge as it affects neonatal survival and influences their normal biological development and metabolism. Several clinical researches have revealed the occurrence of metabolic syndrome, such as insulin resistance, obesity, type 2 diabetes mellitus, oxidative stress, dyslipidemia, as direct results of IUGR. Therefore, it is essential to understand its underlying mechanism, impact and develop effective therapies. The purpose of this work is to review the current knowledge on IUGR induced metabolic syndrome and relevant therapies. Here in, we elaborate on the characteristics and causes of IUGR by pointing out recent research findings. Furthermore, we discuss the impact of IUGR on different organs of the body, followed by preclinical studies on IUGR using suitable animal models. Additionally, various metabolic disorders with their genetic implications, such as insulin resistance, type 2 diabetes mellitus, dyslipidemia, obesity are detailed. Finally, the current therapeutic options used in the treatment of IUGR are summarized with some prospective therapies highlighted.
Collapse
Affiliation(s)
- Alpha Kalonda Mutamba
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaori He
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tao Wang
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, China
| |
Collapse
|
48
|
Wiese MD, Meakin AS, Varcoe TJ, Darby JRT, Sarr O, Kiser P, Bradshaw EL, Regnault TRH, Morrison JL. Hepatic cytochrome P450 function is reduced by life-long Western diet consumption in guinea pig independent of birth weight. Life Sci 2021; 287:120133. [PMID: 34774623 DOI: 10.1016/j.lfs.2021.120133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is characterised by accumulation of triglycerides and cholesterol within the liver and dysregulation of specific hepatic cytochrome P450 (CYPs) activity. CYPs are involved in the metabolism of endogenous and exogenous chemicals. Hepatic CYP activity is dysregulated in human studies and animal models of a Western diet (WD) or low birth weight (LBW) independently, but the additive effects of LBW and postnatal WD consumption are unknown. As such, the aim of this study was to determine the independent and combined effect of birthweight and postnatal diet on hepatic CYP activity in a guinea pig model. METHODS LBW was generated via uterine artery ablation at mid gestation (term = 70 days gestation). Normal birthweight (NBW) and LBW pups were allocated either a control diet (CD) or WD at weaning. After 4 months of dietary intervention, guinea pigs were humanely killed, and liver tissue collected for biochemical and functional hepatic CYP activity analyses. RESULTS Independent of birthweight, functional activity of CYP3A was significantly reduced in female and male WD compared to CD animals (female, P < 0.0001; male, P = 0.004). Likewise, CYP1A2 activity was significantly reduced in male WD compared to CD animals (P = 0.020) but this same reduction was not observed in females. CONCLUSION Diet, but not birthweight, significantly altered hepatic CYP activity in both sexes, and the effect of diet appeared to be greater in males. These findings may have clinical implications for the management of NAFLD and associated co-morbidities between the sexes.
Collapse
Affiliation(s)
- Michael D Wiese
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Ashley S Meakin
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Tamara J Varcoe
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Jack R T Darby
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Ousseynou Sarr
- Departments of Physiology and Pharmacology, Western University, London, ON N6A 5C1, Canada
| | - Patti Kiser
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5C1, Canada
| | - Emma L Bradshaw
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Timothy R H Regnault
- Departments of Physiology and Pharmacology, Western University, London, ON N6A 5C1, Canada; Departments of Obstetrics and Gynaecology, Western University, London, ON N6A 5C1, Canada; Children's Health Research Institute and Lawson Health Research Institute, London, ON N6A 5C1, Canada
| | - Janna L Morrison
- Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
49
|
Gibbs RL, Yates DT. The Price of Surviving on Adrenaline: Developmental Programming Responses to Chronic Fetal Hypercatecholaminemia Contribute to Poor Muscle Growth Capacity and Metabolic Dysfunction in IUGR-Born Offspring. FRONTIERS IN ANIMAL SCIENCE 2021; 2:769334. [PMID: 34966907 PMCID: PMC8713512 DOI: 10.3389/fanim.2021.769334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Maternofetal stress induces fetal programming that restricts skeletal muscle growth capacity and metabolic function, resulting in intrauterine growth restriction (IUGR) of the fetus. This thrifty phenotype aids fetal survival but also yields reduced muscle mass and metabolic dysfunction after birth. Consequently, IUGR-born individuals are at greater lifelong risk for metabolic disorders that reduce quality of life. In livestock, IUGR-born animals exhibit poor growth efficiency and body composition, making these animals more costly and less valuable. Specifically, IUGR-associated programming causes a greater propensity for fat deposition and a reduced capacity for muscle accretion. This, combined with metabolic inefficiency, means that these animals produce less lean meat from greater feed input, require more time on feed to reach market weight, and produce carcasses that are of less quality. Despite the health and economic implications of IUGR pathologies in humans and food animals, knowledge regarding their specific underlying mechanisms is lacking. However, recent data indicate that adaptive programing of adrenergic sensitivity in multiple tissues is a contributing factor in a number of IUGR pathologies including reduced muscle mass, peripheral insulin resistance, and impaired glucose metabolism. This review highlights the findings that support the role for adrenergic programming and how it relates to the lifelong consequences of IUGR, as well as how dysfunctional adrenergic signaling pathways might be effective targets for improving outcomes in IUGR-born offspring.
Collapse
Affiliation(s)
- Rachel L. Gibbs
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Dustin T. Yates
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
50
|
Ren J, Lock MC, Darby JRT, Orgeig S, Holman SL, Quinn M, Seed M, Muhlhausler BS, McMillen IC, Morrison JL. PPARγ activation in late gestation does not promote surfactant maturation in the fetal sheep lung. J Dev Orig Health Dis 2021; 12:963-974. [PMID: 33407953 DOI: 10.1017/s204017442000135x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Respiratory distress syndrome results from inadequate functional pulmonary surfactant and is a significant cause of mortality in preterm infants. Surfactant is essential for regulating alveolar interfacial surface tension, and its synthesis by Type II alveolar epithelial cells is stimulated by leptin produced by pulmonary lipofibroblasts upon activation by peroxisome proliferator-activated receptor γ (PPARγ). As it is unknown whether PPARγ stimulation or direct leptin administration can stimulate surfactant synthesis before birth, we examined the effect of continuous fetal administration of either the PPARγ agonist, rosiglitazone (RGZ; Study 1) or leptin (Study 2) on surfactant protein maturation in the late gestation fetal sheep lung. We measured mRNA expression of genes involved in surfactant maturation and showed that RGZ treatment reduced mRNA expression of LPCAT1 (surfactant phospholipid synthesis) and LAMP3 (marker for lamellar bodies), but did not alter mRNA expression of PPARγ, surfactant proteins (SFTP-A, -B, -C, and -D), PCYT1A (surfactant phospholipid synthesis), ABCA3 (phospholipid transportation), or the PPARγ target genes SPHK-1 and PAI-1. Leptin infusion significantly increased the expression of PPARγ and IGF2 and decreased the expression of SFTP-B. However, mRNA expression of the majority of genes involved in surfactant synthesis was not affected. These results suggest a potential decreased capacity for surfactant phospholipid and protein production in the fetal lung after RGZ and leptin administration, respectively. Therefore, targeting PPARγ may not be a feasible mechanistic approach to promote lung maturation.
Collapse
Affiliation(s)
- Jiaqi Ren
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Hospital for Sick Children, Toronto, ON, Canada
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, 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, SA, Australia
| | - Sandra Orgeig
- Cancer Research Institute, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - 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
| | - Megan Quinn
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mike Seed
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Hospital for Sick Children, Toronto, ON, Canada
| | | | - I Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, 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 Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|