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
|
Flouri D, Darby JRT, Holman SL, Williams G, Vavourakis V, David AL, Morrison JL, Melbourne A. Feasibility of multimodal magnetic resonance imaging to assess maternal hyperoxygenation in sheep pregnancy. J Physiol 2025; 603:1029-1044. [PMID: 39937834 PMCID: PMC11870080 DOI: 10.1113/jp287272] [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: 07/30/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
An adequate supply of oxygen is crucial for optimal fetal growth and development. Estimation of quantitative indices that reflect tissue diffusivity and oxygenation have been enabled by advances in magnetic resonance imaging (MRI) technology. However, the current diagnostic tools in clinical obstetrics, such as Doppler ultrasound measurements of umbilical blood flow and cardiotocography, do not offer direct information about the oxygen supply to the fetus, nor placental function in vivo. Although MRI provides an opportunity to identify critical changes in fetal oxygenation, exact tissue oxygen content cannot be established in humans. Preclinical models such as pregnant sheep allow the use of invasive methods to validate MRI measurements. The present study investigates the relationship between changes in MRI signal and conventional blood gas analyser measurements during normoxic and hyperoxic conditions in pregnant sheep. Several studies have reported an increase in human fetal oxygenation during 100% maternal oxygen inhalation. We investigated the physiological impact of maternal hyperoxygenation on the placenta in normal pregnant sheep using multimodal functional MRI. Using a multicompartment MRI signal model, we observed the expected increase in feto-placental oxygen saturation with maternal hyperoxygenation. In addition, maternal hyperoxygenation resulted in a significant increase in blood-oxygenation-level-dependent (BOLD) signal intensities, suggesting that BOLD MRI allows non-invasive assessment of the feto-placental response to maternal hyperoxygenation in sheep. Our data suggest that diffusion and relaxation-based MRI is sensitive to acute changes in maternal and feto-placental oxygenation and demonstrate a link between MRI-parameter estimated and reference oxygen saturation. KEY POINTS: Quantification of feto-placental oxygenation and function are important for correct differential diagnosis of placental insufficiency. The only current method for obtaining information about fetal oxygen delivery is cordocentesis. However, there is a risk of inducing preterm birth and/or fetal loss associated with the procedure. Magnetic resonance imaging (MRI) can estimate changes in oxygenation in specific areas of placental and fetal tissue. Using the DECIDE (i.e. diffusion-relaxation combined imaging for detailed placental evaluation) multicompartment model that is sensitive to changes in maternal and feto-placental oxygenation and the blood-oxygenation-level-dependent (BOLD) MRI technique in the sheep fetus, we have demonstrated that maternal hyperoxygenation increases oxygenation of fetal tissue in the placenta. There was a differential effect according to placentome morphological type. This study shows a link between MRI estimated parameters and reference maternal and fetalS O 2 ${{S}_{{{{\mathrm{O}}}_2}}}$ andP O 2 ${{P}_{{{{\mathrm{O}}}_2}}}$ by blood gas analyser, supporting the possibility of using multimodal MRI for measuring regional changes in tissue oxygenation in vivo.
Collapse
Affiliation(s)
- Dimitra Flouri
- In Silico Modelling Group, Department of Mechanical & Manufacturing EngineeringUniversity of CyprusNicosiaCyprus
- School of Biomedical Engineering & Imaging SciencesKing's College LondonLondonUK
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical InnovationUniSA: Clinical and Health SciencesAdelaideSAAustralia
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical InnovationUniSA: Clinical and Health SciencesAdelaideSAAustralia
| | - Georgia Williams
- Preclinical Imaging and Research LaboratoriesSouth Australian Health and Medical Research InstituteAdelaideSAAustralia
| | - Vasileios Vavourakis
- In Silico Modelling Group, Department of Mechanical & Manufacturing EngineeringUniversity of CyprusNicosiaCyprus
- Department of Medical Physics & Biomedical EngineeringUniversity College LondonLondonUK
| | - Anna L. David
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonUK
| | - Janna L. Morrison
- Preclinical Imaging and Research LaboratoriesSouth Australian Health and Medical Research InstituteAdelaideSAAustralia
| | - Andrew Melbourne
- School of Biomedical Engineering & Imaging SciencesKing's College LondonLondonUK
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonUK
| |
Collapse
|
3
|
Freedman AA, Miller GE, Franklin AD, Keenan-Devlin LS, Gilman SE, Borders A, Khan SS, Ernst LM. Placental Pathology and Blood Pressure at Age 7: A Longitudinal Discordant Twin Analysis. Arterioscler Thromb Vasc Biol 2025; 45:312-322. [PMID: 39697173 PMCID: PMC11771519 DOI: 10.1161/atvbaha.124.321666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Evidence suggests that the intrauterine environment shapes offspring cardiovascular disease risk. Although placental dysfunction may be an important pathophysiologic pathway, numerous parental and pregnancy characteristics that influence offspring blood pressure are strong confounders of the mechanistic role of the placenta in observational analyses of singletons. Therefore, we leverage twin- and sibling-based comparison designs to determine whether placental pathology is associated with offspring blood pressure at age 7 while mitigating major sources of confounding. METHODS Data are from pregnant participants and their offspring in the Collaborative Perinatal Project, a longitudinal pregnancy cohort conducted from 1959 to 1965 in the United States. After delivery, placentas were systematically examined for lesions indicative of maternal vascular malperfusion (MVM) and acute inflammation. Blood pressure was assessed at a follow-up research visit when the offspring were 7 years old. Linear fixed-effects models were used to estimate associations between within-twin or sibling discordance in placental pathology and differences in blood pressure at age 7. RESULTS Overall, 193 twin pairs were eligible for inclusion, and 23.3% had placentas discordant for MVM. In a fixed-effect analysis, a twin with high-grade MVM had a higher systolic blood pressure Z score by 0.56 SDs than their co-twin without MVM (95% CI, 0.06-1.05) or a 5.7-mm Hg difference (95% CI, 0.6-10.8). Associations were consistent in a sensitivity analysis restricted to dichorionic twins and in a secondary analysis of 759 MVM-discordant sibling pairs. Acute placental inflammation was not associated with blood pressure at age 7. CONCLUSIONS MVM in the placenta is associated with higher offspring blood pressure in mid-childhood, independent of parental and pregnancy characteristics that twins have in common. The findings support the role of the placenta and the intrauterine environment in the developmental origins of cardiovascular health.
Collapse
Affiliation(s)
- Alexa A Freedman
- Department of Obstetrics and Gynecology, Endeavor Health, Evanston, IL (A.A.F.)
- Now with Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (A.A.F.)
| | - Gregory E Miller
- Institute for Policy Research (G.E.M.), Northwestern University, Evanston, IL
- Department of Psychology (G.E.M.), Northwestern University, Evanston, IL
| | - Andrew D Franklin
- Department of Pediatrics, Endeavor Health, Evanston, IL (A.D.F.), Endeavor Health, Evanston, IL
| | - Lauren S Keenan-Devlin
- Department of Obstetrics and Gynecology (L.S.K.-D., A.B.), Endeavor Health, Evanston, IL
- Department of Obstetrics and Gynecology, The University of Chicago Pritzker School of Medicine, Chicago, IL (L.S.K.-D., A.B.)
| | - Stephen E Gilman
- Social and Behavioral Sciences Branch, Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (S.E.G.)
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (S.E.G.)
| | - Ann Borders
- Department of Obstetrics and Gynecology (L.S.K.-D., A.B.), Endeavor Health, Evanston, IL
- Department of Obstetrics and Gynecology, The University of Chicago Pritzker School of Medicine, Chicago, IL (L.S.K.-D., A.B.)
- Department of Medical Social Sciences (A.B.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sadiya S Khan
- Department of Preventive Medicine (S.S.K.), Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Cardiology, Department of Medicine (S.S.K.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine (L.M.E.), Endeavor Health, Evanston, IL
| |
Collapse
|
4
|
Ballasy N, Apantaku I, Dean W, Hemberger M. Off to a good start: The importance of the placental exchange surface - Lessons from the mouse. Dev Biol 2025; 517:248-264. [PMID: 39491740 DOI: 10.1016/j.ydbio.2024.10.009] [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: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The role of the chorio-allantoic placenta as the critical nutrient- and oxygen-supplying organ to nourish the demands of the fetus has been well recognized. This function relies on the successful establishment of the placental feto-maternal exchange unit, or interhaemal barrier, across which all nutrients as well as waste products must pass to cross from the maternal to the fetal blood circulation, or vice versa, respectively. As a consequence, defects in the establishment of this elaborate interface lead to fetal growth retardation or even embryonic lethality, depending on the severity of the defect. Beyond this essential role, however, it has also emerged that the functionality of the feto-maternal interface dictates the proper development of specific embryonic organs, with tightest links observed to the formation of the heart. In this article, we build on the foundational strength of the mouse as experimental model in which the placental causality of embryonic defects can be genetically proven. We discuss in detail the formation of the interhaemal barrier that makes up the labyrinth layer of the murine placenta, including insights into drivers of its formation and the interdependence of the cell types that make up this essential interface, from in vivo and in vitro data using mouse trophoblast stem cells. We highlight mouse genetic tools that enable the elucidation of cause-effect relationships between defects driven by either the trophoblast cells of the placenta or by embryonic cell types. We specifically emphasize gene knockouts for which a placental causality of embryonic heart defects has been demonstrated. This in-depth perspective provides much-needed insights while highlighting remaining gaps in knowledge that are essential for gaining a better understanding of the multi-facetted roles of the placenta in setting us up for a healthy start in life well beyond nutritional support alone.
Collapse
Affiliation(s)
- Noura Ballasy
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Ifeoluwa Apantaku
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Wendy Dean
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Dept. of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Myriam Hemberger
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
5
|
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
|
6
|
Sehgal A, Nold MF, Roberts CT, Menahem S. Vascular responsiveness to low-dose dexamethasone in extremely premature infants: negative influence of fetal growth restriction. Am J Physiol Heart Circ Physiol 2024; 327:H666-H671. [PMID: 39028285 DOI: 10.1152/ajpheart.00375.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/20/2024]
Abstract
Dexamethasone is frequently prescribed for preterm infants to wean from respiratory support and/or to facilitate extubation. This pre-/postintervention prospective study ascertained the impact on clinical (respiratory support) and echocardiographic parameters after dexamethasone therapy in preterm fetal growth restriction (FGR) infants compared with appropriate for gestational age (AGA) infants. Echocardiography was performed within 24 h before the start and after completion of 10-day therapy. Parameters assessed included those reflecting pulmonary vascular resistance and right ventricular output. Seventeen FGR infants (birth gestation and birth weight, 25.2 ± 1.1 wk and 497 ± 92 g, respectively) were compared with 22 AGA infants (gestation and birth weight, 24.5 ± 0.8 and 663 ± 100 g, respectively). Baseline respiratory severity score (mean airway pressure × fractional inspired oxygen) was comparable between the groups, (median [interquartile range] FGR, 10 [6, 13] vs. AGA, 8 ± 2.8, P = 0.08). Pre-dexamethasone parameters of pulmonary vascular resistance (FGR, 0.19 ± 0.03 vs. AGA, 0.2 ± 0.03, P = 0.16) and right ventricular output (FGR, 171 ± 20 vs. 174 ± 17 mL/kg/min, P = 0.6) were statistically comparable. At post-dexamethasone assessments, the decrease in the respiratory severity score was significantly greater in AGA infants (median [interquartile range] FGR, 10 [6, 13] to 9 [2.6, 13.5], P = 0.009 vs. AGA, 8 ± 2.8 to 3 ± 1, P < 0.0001). Improvement in measures of pulmonary vascular resistance (ratio of time to peak velocity to right ventricular ejection time) was greater in AGA infants (FGR, 0.19 ± 0.03 to 0.2 ± 0.03, P = 0.13 vs. AGA 0.2 ± 0.03 to 0.25 ± 0.03, P < 0.0001). The improvement in right ventricular output was significantly greater in AGA infants (171 ± 20 to 190 ± 21, P = 0.014 vs. 174 ± 17 to 203 ± 22, P < 0.0001). This highlights differential cardiorespiratory responsiveness to dexamethasone in extremely preterm FGR infants, which may reflect the in utero maladaptive state.NEW & NOTEWORTHY Dexamethasone (DEX) is frequently used in preterm infants dependent on ventilator support. Differences in vascular structure and function that may have developed prenatally arising from the chronic intrauterine hypoxemia in FGR infants may adversely affect responsiveness. The clinical efficacy of DEX was significantly less in FGR (birth weight < 10th centile) infants, compared with appropriate for gestational age (AGA) infants. Echocardiography showed significantly less improvement in pulmonary vascular resistance in FGR, compared with AGA infants.
Collapse
Affiliation(s)
- Arvind Sehgal
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
- Department of Pediatrics, Monash University, Melbourne, Victoria, Australia
| | - Marcel F Nold
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
- Department of Pediatrics, Monash University, Melbourne, Victoria, Australia
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Calum T Roberts
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
- Department of Pediatrics, Monash University, Melbourne, Victoria, Australia
| | - Samuel Menahem
- Department of Pediatrics, Monash University, Melbourne, Victoria, Australia
- Paediatric and Fetal Cardiac Units, Monash Medical Centre, Monash Health, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Chang EI, Stremming J, Knaub LA, Wesolowski SR, Rozance PJ, Sucharov CC, Reusch JE, Brown LD. Mitochondrial respiration is lower in the intrauterine growth-restricted fetal sheep heart. J Physiol 2024; 602:2697-2715. [PMID: 38743350 PMCID: PMC11325437 DOI: 10.1113/jp285496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
Fetuses affected by intrauterine growth restriction have an increased risk of developing heart disease and failure in adulthood. Compared with controls, late gestation intrauterine growth-restricted (IUGR) fetal sheep have fewer binucleated cardiomyocytes, reflecting a more immature heart, which may reduce mitochondrial capacity to oxidize substrates. We hypothesized that the late gestation IUGR fetal heart has a lower capacity for mitochondrial oxidative phosphorylation. Left (LV) and right (RV) ventricles from IUGR and control (CON) fetal sheep at 90% gestation were harvested. Mitochondrial respiration (states 1-3, LeakOmy, and maximal respiration) in response to carbohydrates and lipids, citrate synthase (CS) activity, protein expression levels of mitochondrial oxidative phosphorylation complexes (CI-CV), and mRNA expression levels of mitochondrial biosynthesis regulators were measured. The carbohydrate and lipid state 3 respiration rates were lower in IUGR than CON, and CS activity was lower in IUGR LV than CON LV. However, relative CII and CV protein levels were higher in IUGR than CON; CV expression level was higher in IUGR than CON. Genes involved in lipid metabolism had lower expression in IUGR than CON. In addition, the LV and RV demonstrated distinct differences in oxygen flux and gene expression levels, which were independent from CON and IUGR status. Low mitochondrial respiration and CS activity in the IUGR heart compared with CON are consistent with delayed cardiomyocyte maturation, and CII and CV protein expression levels may be upregulated to support ATP production. These insights will provide a better understanding of fetal heart development in an adverse in utero environment. KEY POINTS: Growth-restricted fetuses have a higher risk of developing and dying from cardiovascular diseases in adulthood. Mitochondria are the main supplier of energy for the heart. As the heart matures, the substrate preference of the mitochondria switches from carbohydrates to lipids. We used a sheep model of intrauterine growth restriction to study the capacity of the mitochondria in the heart to produce energy using either carbohydrate or lipid substrates by measuring how much oxygen was consumed. Our data show that the mitochondria respiration levels in the growth-restricted fetal heart were lower than in the normally growing fetuses, and the expression levels of genes involved in lipid metabolism were also lower. Differences between the right and left ventricles that are independent of the fetal growth restriction condition were identified. These results indicate an impaired metabolic maturation of the growth-restricted fetal heart associated with a decreased capacity to oxidize lipids postnatally.
Collapse
Affiliation(s)
- Eileen I. Chang
- Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jane Stremming
- Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Leslie A. Knaub
- Department of Medicine, Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Stephanie R. Wesolowski
- Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul J. Rozance
- Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Jane E.B. Reusch
- Department of Medicine, Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Laura D. Brown
- Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
10
|
Malhotra A, Rocha AKAA, Yawno T, Sutherland AE, Allison BJ, Nitsos I, Pham Y, Jenkin G, Castillo-Melendez M, Miller SL. Neuroprotective effects of maternal melatonin administration in early-onset placental insufficiency and fetal growth restriction. Pediatr Res 2024; 95:1510-1518. [PMID: 38225450 PMCID: PMC11126390 DOI: 10.1038/s41390-024-03027-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Early-onset fetal growth restriction (FGR) is associated with adverse outcomes. We hypothesised that maternal melatonin administration will improve fetal brain structure in FGR. METHODS Surgery was performed on twin-bearing ewes at 88 days (0.6 gestation), and FGR induced in one twin via single umbilical artery ligation. Melatonin was administered intravenously (6 mg/day) to a group of ewes commencing on day of surgery until 127 days (0.85 gestation), when the ewe/fetuses were euthanized, and fetal brains collected. RESULTS Study groups were control (n = 5), FGR (n = 5), control+melatonin (control+MLT; n = 6) and FGR+melatonin (FGR + MLT; n = 6). Melatonin administration did not significantly alter fetal body or brain weights. Myelin (CNPase+) fibre density was reduced in FGR vs. control animals in most brain regions examined (p < 0.05) and melatonin treatment restored CNPase fibre density. Similar but less pronounced effect was seen with mature myelin (MBP+) staining. Significant differences in activated microglia (Iba-1) activity were seen between lamb groups (MLT mitigated FGR effect) in periventricular white matter, subventricular zone and external capsule (p < 0.05). Similar effects were seen in astrogliosis (GFAP) in intragyral white matter and cortex. CONCLUSIONS Maternal melatonin administration in early onset FGR led to improved myelination of white matter brain regions, possibly mediated by decreased inflammation. IMPACT Maternal melatonin administration might lead to neuroprotection in the growth-restricted fetus, possibly via dampening neuroinflammation and enhancing myelination. This preclinical study adds to the body of work on this topic, and informs clinical translation. Neuroprotection likely to improve long-term outcomes of this vulnerable infant group.
Collapse
Affiliation(s)
- Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia.
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia.
| | - Anna K A A Rocha
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Li Z, Zhu J, Chen Y, Wei F, Yang J, Tan X. Preeclampsia/eclampsia impacts the structure and function of neonatal hearts probably by reducing myocardial compaction. Eur J Radiol 2024; 173:111382. [PMID: 38382423 DOI: 10.1016/j.ejrad.2024.111382] [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: 11/20/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE Preeclampsia/Eclampsia (PE/E) poses significant risks to neonatal cardiac health. Traditional echocardiographic methods have limitations in detailing these impacts. This study hypothesized that echocardiographic radiomics could provide a more comprehensive assessment of the cardiac changes in neonates affected by PE/E. METHOD In a comprehensive analysis, 2594 neonates underwent echocardiographic screening. From these, 556 were selected for detailed radiomics analysis, focusing on cardiac shape, movement, and texture features. A multiblock sparse partial least squares (sPLS) model integrated these features to assess their association with PE/E. RESULTS Newborns from PE/E-affected pregnancies displayed lower left ventricular ejection fractions compared to the control group (61.1 % vs. 66.2 %). Our radiomics approach extracted 15,494 features per neonate, with the sPLS model identifying 17 features significantly correlated with PE/E. Among these, texture features representing myocardial non-compaction were most strongly correlated with PE/E (correlation coefficient r = 0.63). Detailed visualization of these texture features suggested that PE/E might lead to more pronounced myocardial non-compaction, characterized by a thicker non-compaction layer and increased cardiac trabeculation. CONCLUSIONS Our findings demonstrate the potential of echocardiographic radiomics as a tool for assessing the impact of PE/E on neonatal cardiac function. The correlation between PE/E and myocardial non-compaction underlines the need for enhanced cardiac monitoring in neonates born to PE/E-affected mothers. This study contributes to a better understanding of PE/E's cardiac implications, potentially guiding future clinical practices.
Collapse
Affiliation(s)
- Zexin Li
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, Guangdong 515041, China; Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital), No. 6, Ailong Road, Shenzhen 518172, China
| | - Jinxiu Zhu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, Guangdong 515041, China; Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital), No. 6, Ailong Road, Shenzhen 518172, China
| | - Yequn Chen
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, Guangdong 515041, China
| | - Fengxiang Wei
- Central Laboratory, Longgang District Maternity & Child Healthcare Hospital (Longgang Maternity and Child Institute of Shantou University Medical College), No. 6, Ailong Road, Shenzhen 518172, China
| | - Jinying Yang
- Department of Obstetrics, Longgang District Maternity & Child Healthcare Hospital (Longgang Maternity and Child Institute of Shantou University Medical College), No. 6, Ailong Road, Shenzhen, Guangdong 518172, China
| | - Xuerui Tan
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, Guangdong 515041, China.
| |
Collapse
|
12
|
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
|
13
|
Mahadevan A, Tipler A, Jones H. Shared developmental pathways of the placenta and fetal heart. Placenta 2023; 141:35-42. [PMID: 36604258 DOI: 10.1016/j.placenta.2022.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
Congenital heart defects (CHD) remain the most common class of birth defect worldwide, affecting 1 in every 110 live births. A host of clinical and morphological indicators of placental dysfunction are observed in pregnancies complicated by fetal CHD and, with the recent emergence of single-cell sequencing capabilities, the molecular and physiological associations between the embryonic heart and developing placenta are increasingly evident. In CHD pregnancies, a hostile intrauterine environment may negatively influence and alter fetal development. Placental maldevelopment and dysfunction creates this hostile in-utero environment and may manifest in the development of various subtypes of CHD, with downstream perfusion and flow-related alterations leading to yet further disruption in placental structure and function. The adverse in-utero environment of CHD-complicated pregnancies is well studied, however the specific etiological role that the placenta plays in CHD development remains unclear. Many mouse and rat models have been used to characterize the relationship between CHD and placental dysfunction, but these paradigms present substantial limitations in the assessment of both the heart and placenta. Improvements in non-invasive placental assessment can mitigate these limitations and drive human-specific investigation in relation to fetal and placental development. Here, we review the clinical, structural, and molecular relationships between CHD and placental dysfunction, the CHD subtype-dependence of these changes, and the future of Placenta-Heart axis modeling and investigation.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Alyssa Tipler
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Helen Jones
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA.
| |
Collapse
|
14
|
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
|
15
|
Mattern J, Gemmell A, Allen PE, Mathers KE, Regnault TR, Stansfield BK. Oral pyrroloquinoline quinone (PQQ) during pregnancy increases cardiomyocyte endowment in spontaneous IUGR guinea pigs. J Dev Orig Health Dis 2023; 14:321-324. [PMID: 36861270 PMCID: PMC10202840 DOI: 10.1017/s2040174423000053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) exerts a negative impact on developing cardiomyocytes and emerging evidence suggests activation of oxidative stress pathways plays a key role in this altered development. Here, we provided pregnant guinea pig sows with PQQ, an aromatic tricyclic o-quinone that functions as a redox cofactor antioxidant, during the last half of gestation as a potential antioxidant intervention for IUGR-associated cardiomyopathy. METHODS Pregnant guinea pig sows were randomly assigned to receive PQQ or placebo at mid gestation and fetuses were identified as spontaneous IUGR (spIUGR) or normal growth (NG) near term yielding four cohorts: NG ± PQQ and spIUGR ± PQQ. Cross sections of fetal left and right ventricles were prepared and cardiomyocyte number, collagen deposition, proliferation (Ki67) and apoptosis (TUNEL) were analyzed. RESULTS Cardiomyocyte endowment was reduced in spIUGR fetal hearts when compared to NG; however, PQQ exerted a positive effect on cardiomyocyte number in spIUGR hearts. Cardiomyocytes undergoing proliferation and apoptosis were more common in spIUGR ventricles when compared with NG animals, which was significantly reduced with PQQ supplementation. Similarly, collagen deposition was increased in spIUGR ventricles and was partially rescued in PQQ-treated spIUGR animals. CONCLUSION The negative influence of spIUGR on cardiomyocyte number, apoptosis, and collagen deposition during parturition can be suppressed by antenatal administration of PQQ to pregnant sows. These data identify a novel therapeutic intervention for irreversible spIUGR-associated cardiomyopathy.
Collapse
Affiliation(s)
- Jordan Mattern
- Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Andrew Gemmell
- Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Paige E. Allen
- Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Katherine E. Mathers
- Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Timothy R.H. Regnault
- Departments of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Obstetrics and Gynecology Western University, London, ON, Canada and
- Children’s Health Research Institute, London, ON, Canada
| | | |
Collapse
|
16
|
Freedman AA, Price E, Franklin A, Ernst LM. Measures of Fetal Growth and Cardiac Structure in Stillbirths With Placental Maternal Vascular Malperfusion: Evidence for Heart Weight Sparing and Structural Cardiac Alterations in Humans. Pediatr Dev Pathol 2023:10935266231166548. [PMID: 37082927 DOI: 10.1177/10935266231166548] [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] [Indexed: 04/22/2023]
Abstract
BACKGROUND Placental maternal vascular malperfusion (MVM) is associated with fetal growth restriction (FGR). While FGR increases the risk of cardiovascular disease, the impact of MVM on fetal cardiac structure is understudied. METHODS We utilized a cohort of autopsied stillbirths; 29 with MVM as the cause of death and 21 with a cause of death unrelated to MVM. Fetal and organ weights and heart measurements were standardized by gestational age and compared between MVM and non-MVM stillbirths. Differences in standardized fetal organ and cardiac measures as compared to standardized fetal body weight were calculated to account for body size. RESULTS MVM stillbirths had smaller organ and heart weights than non-MVM stillbirths; however, after accounting for gestational age, heart weight was the least affected among all organs. In an analysis of organ weights relative to body size, heart weights were 0.31 standard deviations (SD) larger than expected relative to body weight (95% CI: 0.04, 0.57). Right and left ventricle thicknesses and mitral valve circumference were also larger than expected relative to body weight. CONCLUSION Stillbirth due to MVM was associated with relative sparing of heart weight and other heart measurements. The significance of these findings in liveborn infants needs further study.
Collapse
Affiliation(s)
- Alexa A Freedman
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Erica Price
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
| | - Andrew Franklin
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
- Department of Pathology, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| |
Collapse
|
17
|
Ramamoorthi Elangovan V, Saadat N, Ghnenis A, Padmanabhan V, Vyas AK. Developmental programming: adverse sexually dimorphic transcriptional programming of gestational testosterone excess in cardiac left ventricle of fetal sheep. Sci Rep 2023; 13:2682. [PMID: 36792653 PMCID: PMC9932081 DOI: 10.1038/s41598-023-29212-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Adverse in-utero insults during fetal life alters offspring's developmental trajectory, including that of the cardiovascular system. Gestational hyperandrogenism is once such adverse in-utero insult. Gestational testosterone (T)-treatment, an environment of gestational hyperandrogenism, manifests as hypertension and pathological left ventricular (LV) remodeling in adult ovine offspring. Furthermore, sexual dimorphism is noted in cardiomyocyte number and morphology in fetal life and at birth. This study investigated transcriptional changes and potential biomarkers of prenatal T excess-induced adverse cardiac programming. Genome-wide coding and non-coding (nc) RNA expression were compared between prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30 to 90 of gestation; Term: 147 days) and control ovine LV at day 90 fetus in both sexes. Prenatal T induced differential expression of mRNAs in the LV of female (2 down, 5 up) and male (3 down, 1 up) (FDR < 0.05, absolute log2 fold change > 0.5); pathways analysis demonstrated 205 pathways unique to the female, 382 unique to the male and 23 common pathways. In the male, analysis of ncRNA showed differential regulation of 15 lncRNAs (14 down, 1 up) and 27 snoRNAs (26 down and 1 up). These findings suggest sexual dimorphic modulation of cardiac coding and ncRNA with gestational T excess.
Collapse
Affiliation(s)
| | - Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Arpita K Vyas
- College of Medicine, California Northstate University, Elk Grove, CA, USA.
- Department of Pediatrics, Division of Pediatric Endocrinology, School of Medicine, Washington University, St Louis, MO, USA.
| |
Collapse
|
18
|
Folguieri MS, Franco ATB, Vieira AS, Gontijo JAR, Boer PA. Transcriptome and morphological analysis on the heart in gestational protein-restricted aging male rat offspring. Front Cell Dev Biol 2022; 10:892322. [PMID: 36353510 PMCID: PMC9638007 DOI: 10.3389/fcell.2022.892322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/10/2022] [Indexed: 09/08/2024] Open
Abstract
Background: Adverse factors that influence embryo/fetal development are correlated with increased risk of cardiovascular disease (CVD), type-2 diabetes, arterial hypertension, obesity, insulin resistance, impaired kidney development, psychiatric disorders, and enhanced susceptibility to oxidative stress and inflammatory processes in adulthood. Human and experimental studies have demonstrated a reciprocal relationship between birthweight and cardiovascular diseases, implying intrauterine adverse events in the onset of these abnormalities. In this way, it is plausible that confirmed functional and morphological heart changes caused by gestational protein restriction could be related to epigenetic effects anticipating cardiovascular disorders and reducing the survival time of these animals. Methods: Wistar rats were divided into two groups according to the protein diet content offered during the pregnancy: a normal protein diet (NP, 17%) or a Low-protein diet (LP, 6%). The arterial pressure was measured, and the cardiac mass, cardiomyocytes area, gene expression, collagen content, and immunostaining of proteins were performed in the cardiac tissue of male 62-weeks old NP compared to LP offspring. Results: In the current study, we showed a low birthweight followed by catch-up growth phenomena associated with high blood pressure development, increased heart collagen content, and cardiomyocyte area in 62-week-old LP offspring. mRNA sequencing analysis identified changes in the expression level of 137 genes, considering genes with a p-value < 0.05. No gene was. Significantly changed according to the adj-p-value. After gene-to-gene biological evaluation and relevance, the study demonstrated significant differences in genes linked to inflammatory activity, oxidative stress, apoptosis process, autophagy, hypertrophy, and fibrosis pathways resulting in heart function disorders. Conclusion: The present study suggests that gestational protein restriction leads to early cardiac diseases in the LP progeny. It is hypothesized that heart dysfunction is associated with fibrosis, myocyte hypertrophy, and multiple abnormal gene expression. Considering the above findings, it may suppose a close link between maternal protein restriction, specific gene expression, and progressive heart failure.
Collapse
Affiliation(s)
- Marina S. Folguieri
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - Ana Teresa Barufi Franco
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas (UNICAMP), Campinas, Brazil
| | - José Antonio Rocha Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - Patricia Aline Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| |
Collapse
|
19
|
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
|
20
|
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]
|
21
|
Starr VJ, Dzialowski EM. Developing chicken cardiac muscle mitochondria are resistant to variations in incubation oxygen levels. Curr Res Physiol 2022; 5:151-157. [PMID: 35345510 PMCID: PMC8956876 DOI: 10.1016/j.crphys.2022.03.001] [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: 12/22/2021] [Revised: 03/05/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Chronic exposure to hypoxia during vertebrate development can produce abnormal cardiovascular morphology and function. The aim of this study was to examine cardiac mitochondria function in an avian model, the chicken, in response to embryonic development under hypoxic (15% O2), normoxic (21% O2), or hyperoxic (40% O2) incubation conditions. Methods Chicken embryos were incubated in hypoxia, normoxia, or hyperoxia beginning on day 5 of incubation through hatching. Cardiac mitochondria oxygen flux and reactive oxygen species production were measured in permeabilized cardiac fibers from externally pipped and 1-day post hatchlings. Results Altering oxygen during development had a large effect on body and heart masses of externally pipped embryos and 1-day old hatchlings. Hypoxic animals had smaller body masses and absolute heart masses, but proportionally similar sized hearts compared to normoxic animals during external pipping. Hyperoxic animals were larger with larger hearts than normoxic animals during external pipping. Mitochondrial oxygen flux in permeabilized cardiac muscle fibers revealed limited effects of developing under altered oxygen conditions, with only oxygen flux through cytochrome oxidase being lower in hypoxic hearts compared with hyperoxic hearts. Oxygen flux in leak and oxidative phosphorylation states were not affected by developmental oxygen levels. Mitochondrial reactive oxygen species production under leak and oxidative phosphorylation states studied did not differ between any developmental oxygen treatment. Conclusions These results suggest that cardiac mitochondria function of the developing chicken is not altered by developing in ovo under different oxygen levels. Chicken heart mass is influenced by oxygen availability during development. Cardiac mitochondria respiration was unchanged by developing under hypoxic or hyperoxic oxygen stress. Cardiac mitochondria ROS production was not altered by developmental oxygen stress.
Collapse
Key Words
- AA, Antimycin A
- ADP, adenosine diphosphate
- COX, cytochrome oxidase
- Cardiac mitochondria
- Chicken
- EP, external pipping
- GMP, glutamate, malate, and pyruvate
- Hyperoxia
- Hypoxia
- IP, internal pipping
- LEAK, mitochondrial leak respiration
- OMY, oligomycin
- OXPHOS, mitochondrial oxidative phosphorylation
- ROS, reactive oxygen species
- ROT, rotenone
- Reactive oxygen species
- S, succinate
- TMPD, N,N,N’,N’-tetramethyl-p-phenylenediamine
- dph, days post hatching
Collapse
Affiliation(s)
- Vanessa J Starr
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| | - Edward M Dzialowski
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| |
Collapse
|
22
|
Sutovska H, Babarikova K, Zeman M, Molcan L. Prenatal Hypoxia Affects Foetal Cardiovascular Regulatory Mechanisms in a Sex- and Circadian-Dependent Manner: A Review. Int J Mol Sci 2022; 23:2885. [PMID: 35270026 PMCID: PMC8910900 DOI: 10.3390/ijms23052885] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 11/17/2022] Open
Abstract
Prenatal hypoxia during the prenatal period can interfere with the developmental trajectory and lead to developing hypertension in adulthood. Prenatal hypoxia is often associated with intrauterine growth restriction that interferes with metabolism and can lead to multilevel changes. Therefore, we analysed the effects of prenatal hypoxia predominantly not associated with intrauterine growth restriction using publications up to September 2021. We focused on: (1) The response of cardiovascular regulatory mechanisms, such as the chemoreflex, adenosine, nitric oxide, and angiotensin II on prenatal hypoxia. (2) The role of the placenta in causing and attenuating the effects of hypoxia. (3) Environmental conditions and the mother's health contribution to the development of prenatal hypoxia. (4) The sex-dependent effects of prenatal hypoxia on cardiovascular regulatory mechanisms and the connection between hypoxia-inducible factors and circadian variability. We identified that the possible relationship between the effects of prenatal hypoxia on the cardiovascular regulatory mechanism may vary depending on circadian variability and phase of the days. In summary, even short-term prenatal hypoxia significantly affects cardiovascular regulatory mechanisms and programs hypertension in adulthood, while prenatal programming effects are not only dependent on the critical period, and sensitivity can change within circadian oscillations.
Collapse
Affiliation(s)
| | | | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia; (H.S.); (K.B.); (L.M.)
| | | |
Collapse
|
23
|
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
|
24
|
Grzęda E, Matuszewska J, Ziarniak K, Gertig-Kolasa A, Krzyśko- Pieczka I, Skowrońska B, Sliwowska JH. Animal Foetal Models of Obesity and Diabetes - From Laboratory to Clinical Settings. Front Endocrinol (Lausanne) 2022; 13:785674. [PMID: 35197931 PMCID: PMC8858803 DOI: 10.3389/fendo.2022.785674] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
The prenatal period, during which a fully formed newborn capable of surviving outside its mother's body is built from a single cell, is critical for human development. It is also the time when the foetus is particularly vulnerable to environmental factors, which may modulate the course of its development. Both epidemiological and animal studies have shown that foetal programming of physiological systems may alter the growth and function of organs and lead to pathology in adulthood. Nutrition is a particularly important environmental factor for the pregnant mother as it affects the condition of offspring. Numerous studies have shown that an unbalanced maternal metabolic status (under- or overnutrition) may cause long-lasting physiological and behavioural alterations, resulting in metabolic disorders, such as obesity and type 2 diabetes (T2DM). Various diets are used in laboratory settings in order to induce maternal obesity and metabolic disorders, and to alter the offspring development. The most popular models are: high-fat, high-sugar, high-fat-high-sugar, and cafeteria diets. Maternal undernutrition models are also used, which results in metabolic problems in offspring. Similarly to animal data, human studies have shown the influence of mothers' diets on the development of children. There is a strong link between the maternal diet and the birth weight, metabolic state, changes in the cardiovascular and central nervous system of the offspring. The mechanisms linking impaired foetal development and adult diseases remain under discussion. Epigenetic mechanisms are believed to play a major role in prenatal programming. Additionally, sexually dimorphic effects on offspring are observed. Therefore, further research on both sexes is necessary.
Collapse
Affiliation(s)
- Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- Molecular and Cell Biology Unit, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Gertig-Kolasa
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Izabela Krzyśko- Pieczka
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogda Skowrońska
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna H. Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- *Correspondence: Joanna H. Sliwowska,
| |
Collapse
|
25
|
McBride GM, Meakin AS, Soo JY, Darby JRT, Varcoe TJ, Bradshaw EL, Lock MC, Holman SL, Saini BS, Macgowan CK, Seed M, Berry MJ, Wiese MD, Morrison JL. Intrauterine growth restriction alters the activity of drug metabolising enzymes in the maternal-placental-fetal unit. Life Sci 2021; 285:120016. [PMID: 34614415 DOI: 10.1016/j.lfs.2021.120016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Ten percent of pregnancies are affected by intrauterine growth restriction (IUGR), and evidence suggests that affected neonates have reduced activity of hepatic cytochrome P450 (CYP) drug metabolising enzymes. Given that almost all pregnant individuals take medications and additional medications are often required during an IUGR pregnancy, we aimed to determine the impact of IUGR on hepatic CYP activity in sheep fetuses and pregnant ewes. METHODS Specific probes were used to determine the impact of IUGR on the activity of several CYP isoenzymes (CYP1A2, CYP2C19, CYP2D6 and CYP3A) in sheep fetuses and pregnant ewes. Probes were administered intravenously to the ewe at 132 days (d) gestation (term 150 d), followed by blood sampling from the maternal and fetal circulation over 24 h. Maternal and fetal liver tissue was collected at 139-140 d gestation, from which microsomes were isolated and incubated with probes. Metabolite and maternal plasma cortisol concentrations were measured using Liquid Chromatography - tandem mass spectrometry (LC-MS/MS). RESULTS Maternal plasma cortisol concentration and maternal hepatic CYP1A2 and CYP3A activity was significantly higher in IUGR pregnancies. Maternal hepatic CYP activity was higher than fetal hepatic CYP activity for all CYPs tested, and there was minimal CYP1A2 or CYP3A activity in the late gestation fetus when assessed using in vitro methods. CONCLUSIONS The physiological changes to the maternal-placental-fetal unit in an IUGR pregnancy have significant effects on maternal drug metabolism, suggesting changes in medications and/or doses may be required to optimise maternal and fetal health.
Collapse
Affiliation(s)
- Grace M McBride
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Ashley S Meakin
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Tamara J Varcoe
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Emma L Bradshaw
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | | | - Brahmdeep S Saini
- The Hospital for Sick Children and University of Toronto, Toronto M5G 1X8, Canada
| | | | - Mike Seed
- The Hospital for Sick Children and University of Toronto, Toronto M5G 1X8, Canada
| | - Mary J Berry
- University of Otago, Wellington, NZ 6242, New Zealand
| | - Michael D Wiese
- Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Australia; Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia.
| |
Collapse
|
26
|
Ferguson DP, Leszczynski EC, McPeek AC, Pendergrast LA, Visker JR, Triplett AN. Physical Activity Engagement Worsens Health Outcomes and Limits Exercise Capacity in Growth-restricted Mice. Med Sci Sports Exerc 2021; 53:1561-1571. [PMID: 34261989 PMCID: PMC10797723 DOI: 10.1249/mss.0000000000002620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION A total of 161 million children a year are growth restricted, leading to a 47% increased risk of chronic disease in adulthood. Physical activity (PA) reduces the risk of mortality from chronic disease. The purpose of the present investigation was to determine the effect of a PA intervention (wheel running) on cardiac and skeletal muscle capacities in gestational (GUN) and postnatal (PUN) growth-restricted mice as compared with nonrestricted controls (CON). METHODS A low-protein cross-fostering FVB mouse model was used to induce growth restriction during gestation and the first 21 d of postnatal life. Mouse pups were recovered on a healthy diet until mature and provided wheel access for 3 wk. At completion of the PA intervention, mice underwent maximal exercise testing on a treadmill, echocardiography, and skeletal muscle histology. RESULTS After the PA intervention, CON mice had a 45% improvement in maximal exercise capacity (P = 0.0390) because of cardiac and skeletal muscle adaptations, but GUN and PUN mice did not. Alarmingly, PUN female mice exposed to wheels had 11.45% lower left ventricular volume (P = 0.0540) and 18% lower left ventricle area (P = 0.0585), with blood flow velocities indicative of cardiac fibrosis (GUN had elevated isovolumetric contraction time P = 0.0374; GUN females and PUN males had longer isovolumetric relaxation time P = 0.0703). PUN male mice had mixed skeletal muscle responses with an oxidative shift in the diaphragm (P = 0.0162) but a glycolytic shift in the extensor digitorum longus (P = 0.0647). PUN female mice had a glycolytic shift in the soleus after wheel running. CONCLUSIONS Unexpectedly, growth-restricted mice were nonresponders to a PA intervention and displayed negative cardiac outcomes.
Collapse
Affiliation(s)
- David P Ferguson
- Department of Kinesiology, Michigan State University, East Lansing, MI
| | | | - Ashley C McPeek
- Department of Kinesiology, Michigan State University, East Lansing, MI
| | | | | | - Ashley N Triplett
- Department of Kinesiology, Michigan State University, East Lansing, MI
| |
Collapse
|
27
|
Placental insufficiency induces a sexually dimorphic response in the expression of cardiac growth and metabolic signalling molecules upon exposure to a postnatal western diet in guinea pigs. J Dev Orig Health Dis 2021; 13:345-357. [PMID: 34308829 DOI: 10.1017/s204017442100043x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
There is a strong relationship between low birth weight (LBW) and an increased risk of developing cardiovascular disease (CVD). In postnatal life, LBW offspring are becoming more commonly exposed to the additional independent CVD risk factors, such as an obesogenic diet. However, how an already detrimentally programmed LBW myocardium responds to a secondary insult, such as an obesogenic diet (western diet; WD), during postnatal life is ill defined. Herein, we aimed to determine in a pre-clinical guinea pig model of CVD, both the independent and interactive effects of LBW and a postnatal WD on the molecular pathways that regulate cardiac growth and metabolism. Uterine artery ablation was used to induce placental insufficiency (PI) in pregnant guinea pigs to generate LBW offspring. Normal birth weight (NBW) and LBW offspring were weaned onto either a Control diet or WD. At ˜145 days after birth (young adulthood), male and female offspring were humanely killed, the heart weighed and left ventricle tissue collected. The mRNA expression of signalling molecules involved in a pathological hypertrophic and fibrotic response was increased in the myocardium of LBW male, but not female offspring, fed a WD as was the mRNA expression of transcription factors involved in fatty acid oxidation. The mRNA expression of glucose transporters was downregulated by LBW and WD in male, but not female hearts. This study has highlighted a sexually dimorphic cardiac pathological hypertrophic and fibrotic response to the secondary insult of postnatal WD consumption in LBW offspring.
Collapse
|
28
|
Sex-dependent vulnerability of fetal nonhuman primate cardiac mitochondria to moderate maternal nutrient reduction. Clin Sci (Lond) 2021; 135:1103-1126. [PMID: 33899910 DOI: 10.1042/cs20201339] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.
Collapse
|
29
|
Darby JRT, Berry MJ, Quinn M, Holman SL, Bradshaw EL, Jesse SM, Haller C, Seed M, Morrison JL. Haemodynamics and cerebral oxygenation of neonatal piglets in the immediate ex utero period supported by mechanical ventilation or ex utero oxygenator. J Physiol 2021; 599:2751-2761. [PMID: 33745149 DOI: 10.1113/jp280803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/15/2021] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS The margin of human viability has extended to the extremes of gestational age (<24 weeks) when the lungs are immature and ventilator-induced lung injury is common. Artificial placenta technology aims to extend gestation ex utero in order to allow the lungs additional time to develop prior to entering an air-breathing environment. We compared the haemodynamics and cerebral oxygenation of piglets in the immediate period post-oxygenator (OXY) transition against both paired in utero measures and uniquely against piglets transitioned onto mechanical ventilation (VENT). Post-transition, OXY piglets became hypotensive with reduced carotid blood flow in comparison with both paired in utero measures and VENT piglets. The addition of a pump to the oxygenator circuit may be required to ensure haemodynamic stability in the immediate post-transition period. ABSTRACT Gestational age at birth is a major predictor of wellbeing; the lower the gestational age, the greater the risk of mortality and morbidity. At the margins of human viability (<24 weeks gestation) immature lungs combined with the need for early ventilatory support means lung injury and respiratory morbidity is common. The abrupt haemodynamic changes consequent on birth may also contribute to preterm-associated brain injury, including intraventricular haemorrhage. Artificial placenta technology aims to support oxygenation, haemodynamic stability and ongoing fetal development ex utero until mature enough to safely transition to a true ex utero environment. We aimed to characterize the impact of birth transition onto either an oxygenator circuit or positive pressure ventilation on haemodynamic and cerebral oxygenation of the neonatal piglet. At 112 days gestation (term = 115 days), fetal pigs underwent instrumentation surgery and transitioned onto either an oxygenator (OXY, n = 5) or ventilatory support (VENT, n = 8). Blood pressure (BP), carotid blood flow and cerebral oxygenation in VENT piglets rose from in utero levels to be significantly higher than OXY piglets post-transition. OXY piglet BP, carotid blood flow and carotid oxygen delivery (DO2 ) decreased from in utero levels post-transition; however, cerebral regional oxygen saturation (rSO2 ) was maintained at fetal-like levels. OXY piglets became hypoxaemic and retained CO2 . Whether OXY piglets are able to maintain cerebral rSO2 under these conditions for a prolonged period is yet to be determined. Improvements to OXY piglet oxygenation may lie in maintaining piglet BP at in utero levels and enhancing oxygenator circuit flow.
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, South Australia, Australia
| | - Mary J Berry
- Centre for Translational Physiology, University of Otago, Wellington, New Zealand.,Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Megan Quinn
- 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
| | - Emma L Bradshaw
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sarah M Jesse
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Christoph Haller
- Univeristy of Toronto and The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mike Seed
- Univeristy of Toronto and The Hospital for Sick Children, 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
| |
Collapse
|
30
|
Clarke GD, Li J, Kuo AH, Moody AJ, Nathanielsz PW. Cardiac magnetic resonance imaging: insights into developmental programming and its consequences for aging. J Dev Orig Health Dis 2021; 12:203-219. [PMID: 33349289 PMCID: PMC7987688 DOI: 10.1017/s2040174420001233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVD) are important consequences of adverse perinatal conditions such as fetal hypoxia and maternal malnutrition. Cardiac magnetic resonance imaging (CMR) can produce a wealth of physiological information related to the development of the heart. This review outlines the current state of CMR technologies and describes the physiological biomarkers that can be measured. These phenotypes include impaired ventricular and atrial function, maladaptive ventricular remodeling, and the proliferation of myocardial steatosis and fibrosis. The discussion outlines the applications of CMR to understanding the developmental pathways leading to impaired cardiac function. The use of CMR, both in animal models of developmental programming and in human studies, is described. Specific examples are given in a baboon model of intrauterine growth restriction (IUGR). CMR offers great potential as a tool for understanding the sequence of dysfunctional adaptations of developmental origin that can affect the human cardiovascular system.
Collapse
Affiliation(s)
- G D Clarke
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - J Li
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - A H Kuo
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - A J Moody
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - P W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
31
|
Silva PS, Hooper HB, Manica E, Merighe GKF, Oliveira SA, Traldi AS, Negrão JA. Heat stress affects the expression of key genes in the placenta, placental characteristics, and efficiency of Saanen goats and the survival and growth of their kids. J Dairy Sci 2021; 104:4970-4979. [PMID: 33551154 DOI: 10.3168/jds.2020-18301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 11/09/2020] [Indexed: 01/18/2023]
Abstract
Heat stress is detrimental during gestation; however, the effects of heat stress on goat placental characteristics and kid survival remain unclear. The objective of this study was to evaluate the effects of heat stress at final gestation on cortisol concentration, placenta characteristics, and the expression of genes related to placenta. Forty-six primiparous and multiparous Saanen goats were subjected to control (CT; under a thermoneutral environment: air temperature between 12°C and 25°C and the relative humidity from 45 to 73%, n = 23) or heat stress (HS; under a climatic chamber: air temperature at 37°C and the relative humidity at 60 to 70% from 0800 to 1600 h, n = 23) from the last 60 d of pregnancy until the first colostrum suckling. The heat challenge imposed on HS goats during the prepartum period increased their rectal temperature, respiratory frequency, and cortisol levels in plasma and amniotic fluid versus CT goats. In the placenta, HS treatment also increased the expression of the HSPA1A gene. Heat-stressed goats also showed significantly lower expression of HSD11B2 and greater expression of MC2R and NR3C1 than CT goats, suggesting that heat stress decreased the effectiveness by which the HSD11B2 enzyme converts cortisol to cortisone and increased placental responsiveness to cortisol. The HS goats took longer to release the placenta with lighter placental cotyledons, and HS goats had a lower ratio between the kid's weight at birth and placenta weight than CT goats. There was no treatment effect on the kids' survival or weights at birth, but the kids from goats subjected to HS presented lesser cortisol concentration and greater mortality rates at weaning than kids from CT goats. Finally, the overexpression of HSPA1A by HS goats suggests a protective response of placenta. However, the heat stress negatively affected the placenta's expulsion length, placental cotyledons number, weight and area, the ratio between kid's weight and placenta weight, and cortisol signaling. Indeed, the upregulation of MC2R and NR3C1 and downregulation of HSD11B2 on placenta caused by heat stress were associated with greater cortisol concentrations in the amniotic fluid of HS goats. Although HS and CT kids had adequate weights and survival rate during the first weeks of life, the heat stress increased the mortality at weaning of HS kids versus CT kids, suggesting that the heat stress effect persists and can change the ability of kids to respond to weaning challenge.
Collapse
Affiliation(s)
- P S Silva
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - H B Hooper
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - E Manica
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - G K F Merighe
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - S A Oliveira
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - A S Traldi
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil
| | - J A Negrão
- Department of Basic Sciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, 13635-900, Brazil.
| |
Collapse
|
32
|
Flouri D, Darby JRT, Holman SL, Perumal SR, David AL, Morrison JL, Melbourne A. Magnetic resonance imaging of placentome development in the pregnant Ewe. Placenta 2021; 105:61-69. [PMID: 33549925 PMCID: PMC7611430 DOI: 10.1016/j.placenta.2021.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Novel imaging measurements of placental development are difficult to validate due to the invasive nature of gold-standard procedures. Animal studies have been important in validation of magnetic resonance imaging (MRI) measurements in invasive preclinical studies, as they allow for controlled experiments and analysis of multiple time-points during pregnancy. This study characterises the longitudinal diffusion and perfusion properties of sheep placentomes using MRI, measurements that are required for future validation studies. METHODS Pregnant ewes were anaesthetised for a MRI session on a 3T scanner. Placental MRI was used to classify placentomes morphologically into three types based on their shape and size at two gestational ages. To validate classification accuracy, placentome type derived from MRI data were compared with placentome categorisation results after delivery. Diffusion-Weighted MRI and T2-relaxometry were used to measure a broad range of biophysical properties of the placentomes. RESULTS MRI morphological classification results showed consistent gestational age changes in placentome shape, as supported by post-delivery gold standard data. The mean apparent diffusion coefficient was significantly higher at 110 days gestation than at late gestation (~140 days; term, 150 days). Mean T2 was higher at mid gestation (152.2 ± 58.1 ms) compared to late gestation (127.8 ms ± 52.0). Significantly higher perfusion fraction was measured in late gestation placentomes that also had a significantly higher fractional anisotropy when compared to the earlier gestational age. DISCUSSION We report baseline measurements of techniques common in placental MRI for the sheep placenta. These measurements are essential to support future validation measurements of placental MRI techniques.
Collapse
Affiliation(s)
- Dimitra Flouri
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, United Kingdom; Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom.
| | - 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, 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, Australia
| | - Sunthara R Perumal
- South Australian Health & Medical Research Institute, Preclinical, Imaging & Research Laboratories, Adelaide, Australia
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, London, United Kingdom; NIHR Biomedical Research Centre, University College London Hospitals, London, United Kingdom
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andrew Melbourne
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, United Kingdom; Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
33
|
Wendel K, Pfeiffer HCV, Fugelseth DM, Nestaas E, Domellöf M, Skålhegg BS, Elgstøen KBP, Rootwelt H, Pettersen RD, Pripp AH, Stiris T, Moltu SJ. Effects of nutrition therapy on growth, inflammation and metabolism in immature infants: a study protocol of a double-blind randomized controlled trial (ImNuT). BMC Pediatr 2021; 21:19. [PMID: 33407269 PMCID: PMC7789285 DOI: 10.1186/s12887-020-02425-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
Background Current nutritional management of infants born very preterm results in significant deficiency of the essential fatty acids (FAs) arachidonic acid (ARA) and docosahexaenoic acid (DHA). The impact of this deficit on brain maturation and inflammation mediated neonatal morbidities are unknown. The aim of this study is to determine whether early supply of ARA and DHA improves brain maturation and neonatal outcomes in infants born before 29 weeks of gestation. Methods Infants born at Oslo University Hospital are eligible to participate in this double-blind randomized controlled trial. Study participants are randomized to receive an enteral FA supplement of either 0.4 ml/kg MCT-oil™ (medium chain triglycerides) or 0.4 ml/kg Formulaid™ (100 mg/kg of ARA and 50 mg/kg of DHA). The FA supplement is given from the second day of life to 36 weeks’ postmenstrual age (PMA). The primary outcome is brain maturation assessed by Magnetic Resonance Imaging (MRI) at term equivalent age. Secondary outcomes include quality of growth, incidence of neonatal morbidities, cardiovascular health and neuro-development. Target sample size is 120 infants (60 per group), this will provide 80% power to detect a 0.04 difference in mean diffusivity (MD, mm2/sec) in major white matter tracts on MRI. Discussion Supplementation of ARA and DHA has the potential to improve brain maturation and reduce inflammation related diseases. This study is expected to provide valuable information for future nutritional guidelines for preterm infants. Trial registration Clinicaltrials.gov ID: NCT03555019. Registered 4 October 2018- Retrospectively registered.
Collapse
Affiliation(s)
- Kristina Wendel
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway.
| | - Helle Cecilie Viekilde Pfeiffer
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway.,Department of Pediatric Neurology, Oslo University Hospital, Oslo, Norway
| | - Drude Merete Fugelseth
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eirik Nestaas
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway.,Department of Pediatrics, Vestfold Hospital Trust, Tønsberg, Norway
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umea University, Umea, Sweden
| | - Bjorn Steen Skålhegg
- Division of Molecular Nutrition, Department of Nutrition, University of Oslo, Oslo, Norway
| | | | - Helge Rootwelt
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Rolf Dagfinn Pettersen
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Tom Stiris
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sissel J Moltu
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
34
|
Darby JRT, Varcoe TJ, Holman SL, McMillen IC, Morrison JL. The reliance on α-adrenergic receptor stimuli for blood pressure regulation in the chronically hypoxaemic fetus is not dependent on post-ganglionic activation. J Physiol 2020; 599:1307-1318. [PMID: 33347615 DOI: 10.1113/jp280693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/30/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Chronic hypoxaemia is associated with intrauterine growth restriction (IUGR) and a predisposition to the development of hypertension in adult life. IUGR fetuses exhibit a greater reliance on α-adrenergic activation for blood pressure regulation. The fetal blood pressure response to post-ganglionic blockade is not different between control and IUGR fetuses. The decrease in mean arterial pressure is greater in the IUGR sheep fetus after α-adrenergic receptor blockade at the level of the vasculature and this is inversely related to fetal P O 2 . The increased reliance that the IUGR fetus has on α-adrenergic activation for maintenance of mean arterial pressure is not a result of increased post-ganglionic sympathetic activation. ABSTRACT Intrauterine growth restriction (IUGR) is associated with an increased risk of cardiovascular disease in adult life. Placental restriction (PR) in sheep results in chronic hypoxaemia and early onset IUGR with increased circulating plasma noradrenaline concentrations. These IUGR fetuses exhibit a greater decrease in mean arterial pressure (MAP) during α-adrenergic blockade. We aimed to determine the role of post-ganglionic sympathetic activation with respect to regulating MAP in IUGR fetal sheep. PR was induced by carunclectomy surgery prior to conception. Fetal vascular catheterization was performed at 110-126 days gestational age (GA) (term, 150 days) in nine control and seven PR-IUGR fetuses. The fetal blood pressure response to both a post-ganglionic and an α-adrenergic receptor blocker was assessed at 116-120 days GA and/or 129-131 days GA. The effect of both post ganglionic and α-adrenergic blockade on fetal blood pressure was then compared between control and IUGR fetuses at both GAs. There was no difference in the effect of post-ganglionic blockade on MAP in control and IUGR fetal sheep at either 116-120 days GA or 129-131 days GA. α-adrenergic receptor blockade decreased MAP to the same extent in both control and IUGR fetuses at 116-120 days GA. At 129-131 days GA, the drop in MAP in response to α-adrenergic receptor blockade was greater in IUGR fetuses than controls. There was a significant inverse relationship between the drop in MAP in response to α-adrenergic receptor blockade at both GAs with fetal P O 2 . Thus, the increased dependence on α-adrenergic activation for blood pressure regulation in the chronically hypoxaemic IUGR fetus is not a result of increased post-ganglionic sympathetic activation.
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
| | - Tamara J Varcoe
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, 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
| | - 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
|
35
|
McBride GM, Soo JY, Varcoe T, Morrison JL, Wiese MD. Development of a method to determine cytochrome P450 1A2, 2C9, 2D6 and 3A4 activity sheep hepatic microsomes. J Pharmacol Toxicol Methods 2020; 106:106934. [PMID: 33080390 DOI: 10.1016/j.vascn.2020.106934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Ex vivo studies of human fetal hepatic drug metabolism are uncommon as it requires access to functional liver tissue and therefore raises practical and ethical concerns. Large animal models provide an alternative opportunity to study changes in cytochrome P450 (CYP) activity in the mother and fetus during pregnancy. We aimed to develop methods to determine the activity of CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in sheep hepatic microsomes. METHODS We identified optimal conditions to determine the activity of CYP1A2 (using the probe drug phenacetin), CYP2C9 (diclofenac), CYP2D6 (dextromethorphan) and CYP3A4 (midazolam) by varying techniques for microsome extraction, probe drug concentration, incubation time and microsome concentration. The specificity of each probe drug was assessed by determining the rate of metabolism when specific CYP enzyme inhibitors were included in the reaction. RESULTS The optimum incubation time and probe drug concentration was six hours with 5 μM phenacetin (CYP1A2), four hours with 10 μM diclofenac (CYP2C9), 30 min with 1 μM of midazolam (CYP3A4) and 10 min with 1 μM dextromethorphan (CYP2D6). For both CYP2D6 and CYP3A4 reactions required 20 μg of microsomal protein, whereas for CYP1A2 and CYP2C9, reactions required 40 μg of microsomal protein. Metabolism of phenacetin, dextromethorphan and midazolam was reduced by specific enzyme inhibitors, but the specific CYP2C9 inhibitor sulfaphenazole did not substantially inhibit diclofenac metabolism. DISCUSSION This study identifies the optimal conditions for determining CYP activity in maternal sheep hepatic microsomes. In doing so, we have developed a standardised protocol for assessment of microsomal activity of CYP3A4, CYP1A2 and CYP2D6, but we were unable to optimise conditions for assessment of CYP2C9. This approach can be applied to investigate the impact of pregnancy complications on maternal and fetal hepatic drug metabolism.
Collapse
Affiliation(s)
- Grace M McBride
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Tamara Varcoe
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Michael D Wiese
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia.
| |
Collapse
|
36
|
Fetal Growth Restriction and Hypertension in the Offspring: Mechanistic Links and Therapeutic Directions. J Pediatr 2020; 224:115-123.e2. [PMID: 32450071 PMCID: PMC8086836 DOI: 10.1016/j.jpeds.2020.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
37
|
Teulings NEWD, Garrud TAC, Niu Y, Skeffington KL, Beck C, Itani N, Conlon FG, Botting KJ, Nicholas LM, Ashmore TJ, Blackmore HL, Tong W, Camm EJ, Derks JB, Logan A, Murphy MP, Ozanne SE, Giussani DA. Isolating adverse effects of glucocorticoids on the embryonic cardiovascular system. FASEB J 2020; 34:9664-9677. [PMID: 32502311 PMCID: PMC7611332 DOI: 10.1096/fj.202000697r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 01/07/2023]
Abstract
Antenatal glucocorticoid therapy reduces mortality in the preterm infant, but evidence suggests off-target adverse effects on the developing cardiovascular system. Whether deleterious effects are direct on the offspring or secondary to alterations in uteroplacental physiology is unclear. Here, we isolated direct effects of glucocorticoids using the chicken embryo, a model system in which the effects on the developing heart and circulation of therapy can be investigated, independent of effects on the mother and/or the placenta. Fertilized chicken eggs were incubated and divided randomly into control (C) or dexamethasone (Dex) treatment at day 14 out of the 21-day incubation period. Combining functional experiments at the isolated organ, cellular and molecular levels, embryos were then studied close to term. Chicken embryos exposed to dexamethasone were growth restricted and showed systolic and diastolic dysfunction, with an increase in cardiomyocyte volume but decreased cardiomyocyte nuclear density in the left ventricle. Underlying mechanisms included a premature switch from tissue accretion to differentiation, increased oxidative stress, and activated signaling of cellular senescence. These findings, therefore, demonstrate that dexamethasone treatment can have direct detrimental off-target effects on the cardiovascular system in the developing embryo, which are independent of effects on the mother and/or placenta.
Collapse
Affiliation(s)
- Noor E. W. D. Teulings
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Tessa A. C. Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Katie L. Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Christian Beck
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Nozomi Itani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Fiona G. Conlon
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kimberley J. Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Lisa M. Nicholas
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Thomas J. Ashmore
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Heather L. Blackmore
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Wen Tong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emily J. Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jan B. Derks
- Department of Perinatal Medicine, University Medical Centre, Utrecht, Netherlands
| | - Angela Logan
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Michael P. Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Susan E. Ozanne
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Dino A. Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
38
|
Cho SKS, Darby JRT, Saini BS, Lock MC, Holman SL, Lim JM, Perumal SR, Macgowan CK, Morrison JL, Seed M. Feasibility of ventricular volumetry by cardiovascular MRI to assess cardiac function in the fetal sheep. J Physiol 2020; 598:2557-2573. [PMID: 32378201 DOI: 10.1113/jp279054] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS The application of fetal cardiovascular magnetic resonance imaging (CMR) to assess fetal cardiovascular physiology and cardiac function through the quantification of ventricular volumes has previously been investigated, but the approach has not yet been fully validated. Ventricular output measurements calculated from heart rate and stroke volumes (SV) of the right and left ventricles measured by ventricular volumetry (VV) exhibited a high level of agreement with phase-contrast (PC) blood flow measurements in the main pulmonary artery and ascending aorta, respectively. Ejection fraction of the right ventricle, which is lower than that of the left ventricle in postnatal subjects, was similar to the left ventricular ejection fraction in the fetus; probably due to the different loading conditions present in the fetal circulation. This study provides evidence to support the reliability of VV in the sheep fetus, providing evidence for its use in animal models of human diseases affecting the fetal circulation. ABSTRACT The application of ventricular volumetry (VV) by cardiovascular magnetic resonance imaging (CMR) in the fetus remains challenging due to the small size of the fetal heart and high heart rate. The reliability of this technique in utero has not yet been established. The aim of this study was to assess the feasibility and reliability of VV in a fetal sheep model of human pregnancy. Right and left ventricular outputs by stroke volume (SV) measured using VV were compared with 2D phase-contrast (PC) CMR measurements of blood flow in the main pulmonary artery (MPA) and ascending aorta (AAo). At 124-140 days (d) gestation, singleton bearing Merino ewes underwent CMR under general anaesthesia using fetal femoral artery catheters, implanted at 109-117d, to trigger cine steady state free precession acquisitions of ventricular short-axis stacks. The short-axis cine stacks were segmented at end-systole and end-diastole, yielding right and left ventricular SV, ejection fraction, and cardiac outputs (SV × heart rate). PC cine acquisitions of MPA and AAo were analysed to measure blood flow, which served as comparators for the right and left cardiac outputs by VV. There was good correlation and agreement between VV and PC measures of ventricular outputs with no significant bias (r2 = 0.926; P < 0.0001; Bias = -4.7 ± 10.5 ml min-1 kg-1 ; 95% limits of agreement: -15.9 to 25.2 ml min-1 kg-1 ). This study validates fetal VV by CMR in a large animal model of human pregnancy and provides preliminary reference values of fetal sheep right and left ventricles in late gestation.
Collapse
Affiliation(s)
- Steven K S Cho
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Early Origins of Adult Health Research Group, School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia.,Division of Cardiology, Hospital for Sick Children, Toronto, Canada
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - Brahmdeep S Saini
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Division of Cardiology, Hospital for Sick Children, Toronto, Canada
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - Jessie Mei Lim
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Cardiology, Hospital for Sick Children, Toronto, Canada
| | - Sunthara Rajan Perumal
- Preclinical, Imaging & Research Laboratories, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Christopher K Macgowan
- Translational Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - Mike Seed
- Division of Cardiology, Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
39
|
Saini BS, Darby JRT, Portnoy S, Sun L, van Amerom J, Lock MC, Soo JY, Holman SL, Perumal SR, Kingdom JC, Sled JG, Macgowan CK, Morrison JL, Seed M. Normal human and sheep fetal vessel oxygen saturations by T2 magnetic resonance imaging. J Physiol 2020; 598:3259-3281. [PMID: 32372463 DOI: 10.1113/jp279725] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/30/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Human fetal Doppler ultrasound and invasive blood gas measurements obtained by cordocentesis or at the time of delivery reveal similarities with sheep (an extensively used model for human fetal cardiovascular physiology). Oxygen saturation (SO2 ) measurements in human fetuses have been limited to the umbilical and scalp vessels, providing little information about normal regional SO2 differences in the fetus. Blood T2 MRI relaxometry presents a non-invasive measure of SO2 in the major fetal vessels. This study presents the first in vivo validation of fetal vessel T2 oximetry against the in vitro T2-SO2 relationship using catheterized sheep fetuses and compares the normal SO2 in the major vessels between the human and sheep fetal circulations. Human fetal vessel SO2 by T2 MRI confirms many similarities with the sheep fetal circulation and is able to demonstrate regional differences in SO2 ; in particular the significantly higher SO2 in the left versus right heart. ABSTRACT Blood T2 magnetic resonance imaging (MRI) relaxometry non-invasively measures oxygen saturation (SO2 ) in major vessels but has not been validated in fetuses in vivo. We compared the blood T2-SO2 relationship in vitro (tubes) and in vivo (vessels) in sheep, and measured SO2 across the normal human and sheep fetal circulations by T2. Singleton pregnant ewes underwent surgery to implant vascular catheters. In vitro and in vivo sheep blood T2 measurements were related to corresponding SO2 measured using a blood gas analyser, as well as relating T2 and SO2 of human fetal blood in vitro. MRI oximetry was performed in the major vessels of 30 human fetuses at 36 weeks (term, 40 weeks) and 10 fetal sheep (125 days; term, 150 days). The fidelity of in vivo fetal T2 oximetry was confirmed through comparison of in vitro and in vivo sheep blood T2-SO2 relationships (P = 0.1). SO2 was similar between human and sheep fetuses, as was the fetal oxygen extraction fraction (human, 33 ± 11%; sheep, 34 ± 7%; P = 0.798). The presence of streaming in the human fetal circulation was demonstrated by the SO2 gradient between the ascending aorta (68 ± 10%) and the main pulmonary artery (49 ± 9%; P < 0.001). Human and sheep fetal vessel MRI oximetry based on T2 is a validated approach that confirms the presence of streaming of umbilical venous blood towards the heart and brain. Streaming is important in ensuring oxygen delivery to these organs and its disruption may have important implications for organ development, especially in conditions such as congenital heart disease and fetal growth restriction.
Collapse
Affiliation(s)
- Brahmdeep S Saini
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, 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, 5000, Australia
| | - Sharon Portnoy
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Liqun Sun
- Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Joshua van Amerom
- Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, 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, South Australia, 5000, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, 5000, 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, 5000, Australia
| | - Sunthara R Perumal
- Preclinical Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5086, Australia
| | - John C Kingdom
- Department of Obstetrics and Gynaecology, Maternal-Fetal Medicine, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, M5G 1E2, Canada
| | - John G Sled
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Christopher K Macgowan
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, 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, 5000, Australia
| | - Mike Seed
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, M5G 1E2, Canada
| |
Collapse
|
40
|
Kumar P, Morton JS, Shah A, Do V, Sergi C, Serrano‐Lomelin J, Davidge ST, Beker D, Levasseur J, Hornberger LK. Intrauterine exposure to chronic hypoxia in the rat leads to progressive diastolic function and increased aortic stiffness from early postnatal developmental stages. Physiol Rep 2020; 8:e14327. [PMID: 31960611 PMCID: PMC6971413 DOI: 10.14814/phy2.14327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM We sought to explore whether fetal hypoxia exposure, an insult of placental insufficiency, is associated with left ventricular dysfunction and increased aortic stiffness at early postnatal ages. METHODS Pregnant Sprague Dawley rats were exposed to hypoxic conditions (11.5% FiO2 ) from embryonic day E15-21 or normoxic conditions (controls). After delivery, left ventricular function and aortic pulse wave velocity (measure of aortic stiffness) were assessed longitudinally by echocardiography from day 1 through week 8. A mixed ANOVA with repeated measures was performed to compare findings between groups across time. Myocardial hematoxylin and eosin and picro-sirius staining were performed to evaluate myocyte nuclear shape and collagen fiber characteristics, respectively. RESULTS Systolic function parameters transiently increased following hypoxia exposure primarily at week 2 (p < .008). In contrast, diastolic dysfunction progressed following fetal hypoxia exposure beginning weeks 1-2 with lower early inflow Doppler velocities, and less of an increase in early to late inflow velocity ratios and annular and septal E'/A' tissue velocities compared to controls (p < .008). As further evidence of altered diastolic function, isovolumetric relaxation time was significantly shorter relative to the cardiac cycle following hypoxia exposure from week 1 onward (p < .008). Aortic stiffness was greater following hypoxia from day 1 through week 8 (p < .008, except week 4). Hypoxia exposure was also associated with altered nuclear shape at week 2 and increased collagen fiber thickness at week 4. CONCLUSION Chronic fetal hypoxia is associated with progressive LV diastolic dysfunction, which corresponds with changes in nuclear shape and collagen fiber thickness, and increased aortic stiffness from early postnatal stages.
Collapse
Affiliation(s)
- Praveen Kumar
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Jude S. Morton
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Amin Shah
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Victor Do
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Consolato Sergi
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonABCanada
| | - Jesus Serrano‐Lomelin
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
| | - Sandra T. Davidge
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Donna Beker
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Jody Levasseur
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Lisa K. Hornberger
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
41
|
Sazonova ЕN, Cimbalist NA, Kaplieva OV, Lebed’ko OA. The influence of non-opiate analogue of leu-enkephalin to the cardiac consequences of intrauterine hypoxia of albino rats. RUSSIAN OPEN MEDICAL JOURNAL 2019. [DOI: 10.15275/rusomj.2019.0401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objective ― Our study aimed to evaluate the possibility of correcting cardiac consequences of intrauterine hypoxia (IUH) by injecting leu-enkephalin analog, lacking affinity for opiate receptors, in the early postnatal period. Material and Methods ― To model IUH, we placed pregnant Wistar rats in a hypobaric chamber with an oxygen partial pressure of 52 mmHg. The procedure was repeated for 4 h daily over the 15th-19th days of gestation. From the 2nd through the 6th days of their lives, the offspring were injected intraperitoneally with non-opiate leu-enkephalin analog at a dose of 100 μg/kg (NALE: Phe-D-Ala-Gly-Phe-Leu-Arg). This analog did not have affinity for opiate receptors. The 7- and 60-day old offspring of female rats subjected to IUH were investigated. The control group included the descendants of intact animals. We investigated gravimetric indicators, DNA-synthetic activity of cardiomyocytes (CMC) by tritium-labeled thymidine autoradiography method, the size of the CMC nuclei, as well as size and amount of nucleoli in the CMC nuclei. The activity of free radical oxidation was evaluated in cardiac homogenates by chemiluminescence. Results ― In 7-day old rats subjected to IUH vs. control animals, we observed decreases in body mass by 32.6%, in heart mass by 27.3%; in the proportion of 3Н-thymidine labeled CMC nuclei by 32.7% in the left ventricle and by 30.4% in the right ventricle; in the number of nucleoli in the CMC nuclei (in the left ventricle: control – 2.384±0.027, IUH – 2.282±0.027*, p<0.05; in the right ventricle: control – 2.409±0.038; IUH – 2.240±0.012*, p<0, 05). Increase in CML indices of cardiac homogenates was revealed, indicating the activation of free radical oxidation. In 7-day old rats subjected to IUH and administration of the NALE peptide from the 2nd through the 6th days of their lives, the proportion of 3H-thymidine labeled nuclei in the CMC did not differ from the control (in the left ventricle: control – 12.79±0.89%, IUH + NALE – 10.98±0.95%, p>0.05; in the right ventricle: control – 11.61±0.78%; IUH + NALE – 11.26±0.58%, p>0.05). The number of nucleoli in the CMC nuclei of the left and right ventricles in the heart of 7-day old animals in the IUH + NALE group did not differ from the control too. The CML indices of heart homogenates in the IUH + NALE group were significantly lower than those in the IUH group. In 60-day old male rats exposed to IUH, there was a decrease in heart mass by 18.5%, sizes of CMC nuclei by 7.5% and 16.1% in the left and right ventricles, respectively, and in the total nucleoli area in the CMC nuclei of the left ventricle (control – 3.953±0.085; IUH – 3.372±0.078*; p<0.05). In 60-day old male rats subjected to IUH and injections of the NALE peptide from the 2nd to the 6th days of their lives, heart mass (control – 692.73±26.81 mg; IUH + NALE – 631.0±29.79 mg; p>0.05) and the size of the CMC nuclei of the right ventricle (control – 54.25±0.84; IUH + NALE – 55.24±0.94; p>0.05) did not differ significantly from the control. The size of the nuclei, the number and size of the nucleoli in the CMC of the left ventricle, as well as the area of the nucleoli in the CMC of the right ventricle in 60-day old male rats of the IUH + NALE group significantly exceeded control group values. Conclusion ― Administration of the NALE peptide to albino rats subjected to IUH normalized DNA-synthetic activity and the number of nucleoli in the nuclei of CMC in 7-day old animals, and also reduced the severity of oxidative stress in the heart tissue. In 60-day old albino male rats exposed to IUH, injecting NALE from the 2nd to the 6th days of their lives eliminated declines in heart mass and sizes of the CMC nuclei and nucleoli, and also led to an increase in the values of the nucleus-and-nucleolus complex indices compared with the control.
Collapse
|
42
|
Chattergoon NN. Thyroid hormone signaling and consequences for cardiac development. J Endocrinol 2019; 242:T145-T160. [PMID: 31117055 PMCID: PMC6613780 DOI: 10.1530/joe-18-0704] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 01/10/2023]
Abstract
The fetal heart undergoes its own growth and maturation stages all while supplying blood and nutrients to the growing fetus and its organs. Immature contractile cardiomyocytes proliferate to rapidly increase and establish cardiomyocyte endowment in the perinatal period. Maturational changes in cellular maturation, size and biochemical capabilities occur, and require, a changing hormonal environment as the fetus prepares itself for the transition to extrauterine life. Thyroid hormone has long been known to be important for neuronal development, but also for fetal size and survival. Fetal circulating 3,5,3'-triiodothyronine (T3) levels surge near term in mammals and are responsible for maturation of several organ systems, including the heart. Growth factors like insulin-like growth factor-1 stimulate proliferation of fetal cardiomyocytes, while thyroid hormone has been shown to inhibit proliferation and drive maturation of the cells. Several cell signaling pathways appear to be involved in this complicated and coordinated process. The aim of this review was to discuss the foundational studies of thyroid hormone physiology and the mechanisms responsible for its actions as we speculate on potential fetal programming effects for cardiovascular health.
Collapse
Affiliation(s)
- Natasha N Chattergoon
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
43
|
Agnew EJ, Garcia-Burgos A, Richardson RV, Manos H, Thomson AJW, Sooy K, Just G, Homer NZM, Moran CM, Brunton PJ, Gray GA, Chapman KE. Antenatal dexamethasone treatment transiently alters diastolic function in the mouse fetal heart. J Endocrinol 2019; 241:279-292. [PMID: 31013474 PMCID: PMC6541236 DOI: 10.1530/joe-18-0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022]
Abstract
Endogenous glucocorticoid action is important in the structural and functional maturation of the fetal heart. In fetal mice, although glucocorticoid concentrations are extremely low before E14.5, glucocorticoid receptor (GR) is expressed in the heart from E10.5. To investigate whether activation of cardiac GR prior to E14.5 induces precocious fetal heart maturation, we administered dexamethasone in the drinking water of pregnant dams from E12.5 to E15.5. To test the direct effects of glucocorticoids upon the cardiovascular system we used SMGRKO mice, with Sm22-Cre-mediated disruption of GR in cardiomyocytes and vascular smooth muscle. Contrary to expectations, echocardiography showed no advancement of functional maturation of the fetal heart. Moreover, litter size was decreased 2 days following cessation of antenatal glucocorticoid exposure, irrespective of fetal genotype. The myocardial performance index and E/A wave ratio, markers of fetal heart maturation, were not significantly affected by dexamethasone treatment in either genotype. Dexamethasone treatment transiently decreased the myocardial deceleration index (MDI; a marker of diastolic function), in control fetuses at E15.5, with recovery by E17.5, 2 days after cessation of treatment. MDI was lower in SMGRKO than in control fetuses and was unaffected by dexamethasone. The transient decrease in MDI was associated with repression of cardiac GR in control fetuses following dexamethasone treatment. Measurement of glucocorticoid levels in fetal tissue and hypothalamic corticotropin-releasing hormone (Crh) mRNA levels suggest complex and differential effects of dexamethasone treatment upon the hypothalamic-pituitary-adrenal axis between genotypes. These data suggest potentially detrimental and direct effects of antenatal glucocorticoid treatment upon fetal heart function.
Collapse
Affiliation(s)
- E J Agnew
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A Garcia-Burgos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - R V Richardson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - H Manos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A J W Thomson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K Sooy
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - G Just
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - N Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - C M Moran
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - P J Brunton
- Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, UK
| | - G A Gray
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K E Chapman
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
44
|
Sawyer AA, Pollock NK, Gutin B, Weintraub NL, Stansfield BK. Proportionality at birth and left ventricular hypertrophy in healthy adolescents. Early Hum Dev 2019; 132:24-29. [PMID: 30953878 PMCID: PMC7101490 DOI: 10.1016/j.earlhumdev.2019.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Perinatal growth has important implications for cardiac development. Low birth weight is associated with cardiovascular (CV) events and mortality, and animal studies have shown that fetal growth restriction is associated with cardiac remodeling in the perinatal period leading to a permanent loss of cardiomyocyte endowment and compensatory hypertrophy. AIMS To determine associations of birthweight (BW) and multiple proportionality indexes (body mass index (BMI); weight/length2 and Ponderal index (PI); weight/length3) at birth on one hand, with left ventricular (LV) structure and function during adolescence. SUBJECTS 379 healthy adolescents aged 14-18 years in Augusta, Georgia. OUTCOME MEASURES LV structure and function parameters, including intraventricular septal thickness in diastole (IVSd), LV internal dimension in diastole (LVIDd), LV internal diameter in systole (LVIDs), LV posterior wall thickness in diastole (LVPWd), relative wall thickness (RWT), midwall fractional shortening (MFS), and ejection fraction, were assessed by echocardiography. RESULTS When associations of birthweight, birth BMI, and birth PI with LV structure and function parameters were separately evaluated with linear regression adjusting for age, sex, race, Tanner stage, socioeconomic status, and physical activity, significant positive associations of BW with LVIDd (P = 0.004), birth BMI with LV mass index (P = 0.01), and birth PI with IVSd (P = 0.02), LVPWd (P = 0.03), and LV mass index (P = 0.002) were identified. When LV structure and function parameters were compared across PI tertiles, a significant U-shaped trend for LV mass index (Pquadratic = 0.04) was identified. CONCLUSIONS Our adolescent data suggest that proportionality at birth may identify associations between perinatal growth and cardiac remodeling independent of birthweight alone.
Collapse
Affiliation(s)
- Alexandra A. Sawyer
- Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Norman K. Pollock
- Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, USA,Georgia Prevention Institute, Augusta University, Augusta, GA, USA
| | - Bernard Gutin
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Neal L. Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA,Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Brian K. Stansfield
- Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, USA,Vascular Biology Center, Augusta University, Augusta, GA, USA,Corresponding author at: Division of Neonatology, Department of Pediatrics, Medical College of Georgia at Augusta University, 1120 15th Street, BIW-6033, Augusta, GA 30912, USA. (B.K. Stansfield)
| |
Collapse
|
45
|
Ferguson DP, Monroe TO, Heredia CP, Fleischmann R, Rodney GG, Taffet GE, Fiorotto ML. Postnatal undernutrition alters adult female mouse cardiac structure and function leading to limited exercise capacity. J Physiol 2019; 597:1855-1872. [PMID: 30730556 DOI: 10.1113/jp277637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/01/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Impaired growth during fetal life can reprogramme heart development and increase the risk for long-term cardiovascular dysfunction. It is uncertain if the developmental window during which the heart is vulnerable to reprogramming as a result of inadequate nutrition extends into the postnatal period. We found that adult female mice that had been undernourished only from birth to 3 weeks of age had disproportionately smaller hearts compared to males, with thinner ventricle walls and more mononucleated cardiomyocytes. In females, but not males, cardiac diastolic function, and heart rate responsiveness to adrenergic stimulation were limited and maximal exercise capacity was compromised. These data suggest that the developmental window during which the heart is vulnerable to reprogramming by inadequacies in nutrient intake may extend into postnatal life and such individuals could be at increased risk for a cardiac event as a result of strenuous exercise. ABSTRACT Adults who experienced undernutrition during critical windows of development are at increased risk for cardiovascular disease. The contribution of cardiac function to this increased disease risk is uncertain. We evaluated the effect of a short episode of postnatal undernutrition on cardiovascular function in mice at the whole animal, organ, and cellular levels. Pups born to control mouse dams were suckled from birth to postnatal day (PN) 21 on dams fed either a control (20% protein) or a low protein (8% protein) isocaloric diet. After PN21 offspring were fed the same control diet until adulthood. At PN70 V ̇ O 2 , max was measured by treadmill test. At PN80 cardiac function was evaluated by echocardiography and Doppler analysis at rest and following β-adrenergic stimulation. Isolated cardiomyocyte nucleation and Ca2+ transients (with and without β-adrenergic stimulation) were measured at PN90. Female mice that were undernourished and then refed (PUN), unlike male mice, had disproportionately smaller hearts and their exercise capacity, cardiac diastolic function, and heart rate responsiveness to adrenergic stimulation were limited. A reduced left ventricular end diastolic volume, impaired early filling, and decreased stored energy at the beginning of diastole contributed to these impairments. Female PUN mice had more mononucleated cardiomyocytes; under resting conditions binucleated cells had a functional profile suggestive of increased basal adrenergic activation. Thus, a brief episode of early postnatal undernutrition in the mouse can produce persistent changes to cardiac structure and function that limit exercise/functional capacity and thereby increase the risk for the development of a wide variety of cardiovascular morbidities.
Collapse
Affiliation(s)
- David P Ferguson
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Kinesiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Tanner O Monroe
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Celia Pena Heredia
- Section of Geriatrics, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ryan Fleischmann
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - George E Taffet
- Section of Geriatrics, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marta L Fiorotto
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
46
|
Morrison JL, Berry MJ, Botting KJ, Darby JRT, Frasch MG, Gatford KL, Giussani DA, Gray CL, Harding R, Herrera EA, Kemp MW, Lock MC, McMillen IC, Moss TJ, Musk GC, Oliver MH, Regnault TRH, Roberts CT, Soo JY, Tellam RL. Improving pregnancy outcomes in humans through studies in sheep. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1123-R1153. [PMID: 30325659 DOI: 10.1152/ajpregu.00391.2017] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Experimental studies that are relevant to human pregnancy rely on the selection of appropriate animal models as an important element in experimental design. Consideration of the strengths and weaknesses of any animal model of human disease is fundamental to effective and meaningful translation of preclinical research. Studies in sheep have made significant contributions to our understanding of the normal and abnormal development of the fetus. As a model of human pregnancy, studies in sheep have enabled scientists and clinicians to answer questions about the etiology and treatment of poor maternal, placental, and fetal health and to provide an evidence base for translation of interventions to the clinic. The aim of this review is to highlight the advances in perinatal human medicine that have been achieved following translation of research using the pregnant sheep and fetus.
Collapse
Affiliation(s)
- Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Kimberley J Botting
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Martin G Frasch
- Department of Obstetrics and Gynecology, University of Washington , Seattle, Washington
| | - Kathryn L Gatford
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Clint L Gray
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash University , Clayton, Victoria , Australia
| | - Emilio A Herrera
- Pathophysiology Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile , Santiago , Chile
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, University of Western Australia , Perth, Western Australia , Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynaecology, Monash University , Clayton, Victoria , Australia
| | - Gabrielle C Musk
- Animal Care Services, University of Western Australia , Perth, Western Australia , Australia
| | - Mark H Oliver
- Liggins Institute, University of Auckland , Auckland , New Zealand
| | - Timothy R H Regnault
- Department of Obstetrics and Gynecology and Department of Physiology and Pharmacology, Western University, and Children's Health Research Institute , London, Ontario , Canada
| | - Claire T Roberts
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
47
|
Jonker SS, Louey S, Roselli CE. Cardiac myocyte proliferation and maturation near term is inhibited by early gestation maternal testosterone exposure. Am J Physiol Heart Circ Physiol 2018; 315:H1393-H1401. [PMID: 30095996 PMCID: PMC6297822 DOI: 10.1152/ajpheart.00314.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Polycystic ovary syndrome is a complex and common disorder in women, and those affected experience an increased burden of cardiovascular disease. It is an intergenerational syndrome, as affected women with high androgen levels during pregnancy "program" fetal development, leading to a similar phenotype in their female offspring. The effect of excess maternal testosterone exposure on fetal cardiomyocyte growth and maturation is unknown. Pregnant ewes received biweekly injections of vehicle (control) or 100 mg testosterone propionate between 30 and 59 days of gestation (early T) or between 60 and 90 days of gestation (late T). Fetuses were delivered at ~135 days of gestation, and their hearts were enzymatically dissociated to measure cardiomyocyte growth (dimensional measurements), maturation (proportion binucleate), and proliferation (nuclear Ki-67 protein). Early T depressed serum insulin-like growth factor 1 and caused intrauterine growth restriction (IUGR; P < 0.0005). Hearts were smaller with early T ( P < 0.001) due to reduced cardiac myocyte maturation ( P < 0.0005) and proliferation ( P = 0.017). Maturation was also lower in male than female fetuses ( P = 0.004) independent of treatment. Late T did not affect cardiac growth. Early excess maternal testosterone exposure depresses circulating insulin-like growth factor 1 near term and causes IUGR in both female and male offspring. These fetuses have small, immature hearts with reduced proliferation, which may reduce cardiac myocyte endowment and predispose to adverse cardiac growth in postnatal life. While excess maternal testosterone exposure leads to polycystic ovary syndrome and cardiovascular disease in female offspring, it may also predispose to complications of IUGR and cardiovascular disease in male offspring. NEW & NOTEWORTHY Using measurements of cardiac myocyte growth and maturation in an ovine model of polycystic ovary syndrome, this study demonstrates that early gestation excess maternal testosterone exposure reduces near-term cardiomyocyte proliferation and maturation in intrauterine growth-restricted female and male fetuses. The effect of testosterone is restricted to exposure during a specific period early in pregnancy, and the effects appear mediated through reduced insulin-like growth factor 1 signaling. Furthermore, male fetuses, regardless of treatment, had fewer mature cardiomyocytes than female fetuses.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Center for Developmental Health, Oregon Health & Science University , Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Samantha Louey
- Center for Developmental Health, Oregon Health & Science University , Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Charles E Roselli
- Department of Physiology and Pharmacology, Oregon Health & Science University , Portland, Oregon
| |
Collapse
|
48
|
Jonker SS, Kamna D, LoTurco D, Kailey J, Brown LD. IUGR impairs cardiomyocyte growth and maturation in fetal sheep. J Endocrinol 2018; 239:253-265. [PMID: 30143557 PMCID: PMC6510659 DOI: 10.1530/joe-18-0382] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/15/2018] [Accepted: 08/24/2018] [Indexed: 12/13/2022]
Abstract
Placental insufficiency causes intrauterine growth restriction (IUGR), a common complication of pregnancy. In skeletal muscle, IUGR reduces fetal myofibril size, reduces myoblast proliferation and reduces expression of genes in cell cycle regulation clusters. The myocardium is striated like skeletal muscle, and IUGR also reduces cell cycle activity and maturation in cardiomyocytes, despite cardiac output preferentially directed to the coronary circulation. We hypothesized that cardiomyocyte growth restriction would be accompanied by similar changes in cell cycle regulation genes and would reduce cardiomyocyte cell cycle activity, number, maturity and size. Pregnant ewes were housed in elevated ambient temperatures from ~40 to ~115 days of gestation (dGA) to produce placental insufficiency and IUGR; fetal hearts were studied at ~134 dGA. Hearts were biopsied for mRNA analysis and then dissociated into individual myocytes (Control n = 8; IUGR n = 15) or dissected (Control n = 9; IUGR n = 13). IUGR fetuses had low circulating insulin and insulin-like growth factor 1 (IGF1) and high circulating cortisol. Bodies and hearts of IUGR fetuses were lighter than those of Controls. Cardiomyocytes of IUGR fetuses were smaller, less mature, less active in the cell cycle and less numerous than in Controls. Further, there was a pattern of downregulation of cell cycle genes in IUGR ventricles. IUGR growth profiles in heart and skeletal muscle suggest similar regulation despite differences in blood and nutrient delivery prioritization. IGF1 signaling is suggested as a mechanism regulating altered growth in IUGR striated muscle and a potential therapeutic candidate.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Daniel Kamna
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Dan LoTurco
- Department of Pediatrics, Perinatal Research Center, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Jenai Kailey
- Department of Pediatrics, Perinatal Research Center, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Laura D Brown
- Department of Pediatrics, Perinatal Research Center, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
49
|
Thompson LP, Chen L, Polster BM, Pinkas G, Song H. Prenatal hypoxia impairs cardiac mitochondrial and ventricular function in guinea pig offspring in a sex-related manner. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1232-R1241. [PMID: 30365351 DOI: 10.1152/ajpregu.00224.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Adverse intrauterine conditions cause fetal growth restriction and increase the risk of adult cardiovascular disease. We hypothesize that intrauterine hypoxia impairs fetal heart function, is sustained after birth, and manifests as both cardiac and mitochondrial dysfunction in offspring guinea pigs (GPs). Pregnant GPs were exposed to 10.5% O2 (HPX) at 50 days of gestation (full term = 65 days) or normoxia (NMX) for the duration of the pregnancy. Pups were allowed to deliver vaginally and raised in a NMX environment. At 90 days of age, mean arterial pressure (MAP) was measured in anesthetized GPs. NMX and prenatally HPX offspring underwent echocardiographic imaging for in vivo measurement of left ventricular cardiac morphology and function, and O2 consumption rates and complex IV enzyme activity were measured from isolated cardiomyocytes and mitochondria, respectively. Prenatal HPX increased ( P < 0.01) MAP (52.3 ± 1.3 and 58.4 ± 1.1 mmHg in NMX and HPX, respectively) and decreased ( P < 0.05) stroke volume (439.8 ± 54.5 and 289.4 ± 15.8 μl in NMX and HPX, respectively), cardiac output (94.4 ± 11.2 and 67.3 ± 3.8 ml/min in NMX and HPX, respectively), ejection fraction, and fractional shortening in male, but not female, GPs. HPX had no effect on left ventricular wall thickness or end-diastolic volume in either sex. HPX reduced mitochondrial maximal respiration and respiratory reserve capacity and complex IV activity rates in hearts of male, but not female, GPs. Prenatal HPX is a programming stimulus that increases MAP and decreases cardiac and mitochondrial function in male offspring. Sex-related differences in the contractile and mitochondrial responses suggest that female GPs are protected from cardiovascular programming of prenatal HPX.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Ling Chen
- Department of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Brian M Polster
- Department of Anesthesiology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Gerard Pinkas
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Hong Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
50
|
Wang Q, Zhu C, Sun M, Maimaiti R, Ford SP, Nathanielsz PW, Ren J, Guo W. Maternal obesity impairs fetal cardiomyocyte contractile function in sheep. FASEB J 2018; 33:2587-2598. [PMID: 30289749 DOI: 10.1096/fj.201800988r] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity is a major public health problem worldwide. In the United States, one-third of women of reproductive age are obese. Human studies show that maternal obesity (MO) predisposes offspring to cardiovascular disease. However, the underlying mechanisms remain unclear. Given the similarities between pregnancy in sheep and humans, we studied sheep to examine the impact of MO on fetal cardiomyocyte contractility at term. We observed that MO impaired cardiomyocyte contractility by reducing peak shortening and shortening/relengthening velocity, prolonging time to relengthening. MO disrupted Ca2+ homeostasis in fetal cardiomyocytes, increasing intracellular Ca2+ and inducing cellular Ca2+ insensitivity. The Ca2+-release channel was impaired, but Ca2+ uptake was unaffected by MO. The upstream kinases that phosphorylate the Ca2+-release channel-ryanodine receptor-2, PKA, and calmodulin-dependent protein kinase II-were activated in MO fetuses. Contractile dysfunction was associated with an increased ratio of myosin heavy chain (MHC)-β to MHC-α and upregulated cardiac troponin (cTn)-T and tropomyosin, as well as cTn-I phosphorylation. In summary, this is the first characterization of the effects of MO on fetal cardiomyocyte contractility. Our findings indicate that MO impairs fetal cardiomyocyte contractility through altered intracellular Ca2+ handling, overloading fetal cardiomyocyte intracellular Ca2+ and aberrant myofilament protein composition. These mechanisms may contribute to developmental programming by MO of offspring cardiac function and predisposition to later life cardiovascular disease in the offspring.-Wang, Q., Zhu, C., Sun, M., Maimaiti, R., Ford, S. P., Nathanielsz, P. W., Ren, J., Guo, W. Maternal obesity impairs fetal cardiomyocyte contractile function in sheep.
Collapse
Affiliation(s)
- Qiurong Wang
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Chaoqun Zhu
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Mingming Sun
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Rexiati Maimaiti
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Stephen P Ford
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Peter W Nathanielsz
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming, USA
| | - Wei Guo
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|