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
|
Vrselja A, Pillow JJ, Bensley JG, Ahmadi‐Noorbakhsh S, Noble PB, Black MJ. Dose-related cardiac outcomes in response to postnatal dexamethasone treatment in premature lambs. Anat Rec (Hoboken) 2025; 308:1214-1228. [PMID: 36924351 PMCID: PMC11889478 DOI: 10.1002/ar.25202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Postnatal corticosteroids are used in the critical care of preterm infants for the prevention and treatment of bronchopulmonary dysplasia. We aimed to investigate the effects of early postnatal dexamethasone therapy and dose on cardiac maturation and morphology in preterm lambs. METHODS Lambs were delivered prematurely at ~128 days of gestational age and managed postnatally according to best clinical practice. Preterm lambs were administered dexamethasone daily at either a low-dose (n = 9) or a high-dose (n = 7), or were naïve to steroid treatment and administered saline (n = 9), over a 7-day time-course. Hearts were studied at postnatal Day 7 for gene expression and assessment of myocardial structure. RESULTS High-dose dexamethasone treatment in the early postnatal period led to marked differences in cardiac gene expression, altered cardiomyocyte maturation and reduced cardiomyocyte endowment in the right ventricle, as well as increased inflammatory infiltrates into the left ventricle. Low-dose exposure had minimal effects on the preterm heart. CONCLUSION Neonatal dexamethasone treatment led to adverse effects in the preterm heart in a dose-dependent manner within the first week of life. The observed cardiac changes associated with high-dose postnatal dexamethasone treatment may influence postnatal growth and remodeling of the preterm heart and subsequent long-term cardiac function.
Collapse
Affiliation(s)
- Amanda Vrselja
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Jennifer Jane Pillow
- School of Human SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Jonathan G. Bensley
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | | | - Peter B. Noble
- School of Human SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Mary Jane Black
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
3
|
Amanollahi R, Holman SL, Bertossa MR, Meakin AS, Thornburg KL, McMillen IC, Wiese MD, Lock MC, Morrison JL. Ontogeny of Fetal Cardiometabolic Pathways: The Potential Role of Cortisol and Thyroid Hormones in Driving the Transition from Preterm to Near-Term Heart Development in Sheep. J Cardiovasc Dev Dis 2025; 12:36. [PMID: 39997470 PMCID: PMC11856455 DOI: 10.3390/jcdd12020036] [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: 12/10/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Understanding hormonal and molecular changes during the transition from preterm to near-term gestation is essential for investigating how pregnancy complications impact fetal heart development and contribute to long-term cardiovascular risks for offspring. This study examines these cardiac changes in fetal sheep, focusing on the changes between 116 days (preterm) and 140 days (near term) of gestation (dG, term = 150) using Western blotting, LC-MS/MS, and histological techniques. We observed a strong correlation between cortisol and T3 (Triiodothyronine) in heart tissue in near-term fetuses, highlighting the role of glucocorticoid signalling in fetal heart maturation. Protein expression patterns in the heart revealed a decrease in multiple glucocorticoid receptor isoforms (GRα-A, GR-P, GR-A, GRα-D2, and GRα-D3), alongside a decrease in IGF-1R (a marker of cardiac proliferative capacity) and p-FOXO1(Thr24) but an increase in PCNA (a marker of DNA replication), indicating a shift towards cardiomyocyte maturation from preterm to near term. The increased expression of proteins regulating mitochondrial biogenesis and OXPHOS complex 4 reflects the known transition from glycolysis to oxidative phosphorylation, essential for meeting the energy demands of the postnatal heart. We also found altered glucose transporter expression, with increased pIRS-1(ser789) and GLUT-4 but decreased GLUT-1 expression, suggesting improved insulin responsiveness as the heart approaches term. Notably, the reduced protein abundance of SIRT-1 and SERCA2, along with increased phosphorylation of cardiac Troponin I(Ser23/24), indicates adaptations for more energy-efficient contraction in the near-term heart. In conclusion, these findings show the complex interplay of hormonal, metabolic, and growth changes that regulate fetal heart development, providing new insights into heart development that are crucial for understanding pathological conditions at birth and throughout life.
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, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - 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 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Melanie R. Bertossa
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - 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, OR 97239, 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, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences University of South Australia, Adelaide, SA 5001, 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, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - 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 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| |
Collapse
|
4
|
Dimasi CG, Darby JR, Holman SL, Quinn M, Meakin AS, Seed M, Wiese MD, Morrison JL. Cardiac growth patterns and metabolism before and after birth in swine: Role of miR in proliferation, hypertrophy and metabolism. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100084. [PMID: 39803591 PMCID: PMC11708124 DOI: 10.1016/j.jmccpl.2024.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 01/06/2025]
Abstract
The adult mammalian heart is unable to undergo cardiac repair, limiting potential treatment options after cardiac damage. However, the fetal heart is capable of cardiac repair. In preparation for birth, cardiomyocytes (CMs) undergo major maturational changes that include exit from the cell cycle, hypertrophic growth, and mitochondrial maturation. The timing and regulation of such events in large mammals is not fully understood. In the present study, we aimed to assess this critical CM transition period using pigs as a preclinically relevant model. Left ventricular myocardium from Large White cross Landrace gilts was collected at 91, 98, 106 and 111-113 days gestation (d GA; term = 115d GA) and in piglets at 0-1, 4-5, 14-18, 19-20 days after birth. We found that miR-133a, which has known roles in CM proliferation, was significantly downregulated before birth, before rising postnatally. Likewise, gene expression of PCNA and CDK1 was repressed until birth with a rise postnatally, suggesting a decline in proliferation during late gestation followed by the onset of multinucleation in postnatal life. The timing of the switch in myocardial metabolism was unclear; however, complexes within the electron transport chain and mitochondrial biogenesis followed a similar pattern of decreasing abundance during late gestation and then a rise postnatally. These data suggest that CM maturation events such as cell cycle arrest and mitochondrial maturation occur around birth. These results may prove important to consider for preclinical applications such as the development of new therapeutics for cardiac repair.
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, SA 5001, Australia
| | - Jack R.T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, 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 5001, Australia
| | - Megan Quinn
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, 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, SA 5001, Australia
| | - Mike Seed
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Division of Cardiology, Toronto, ON, Canada
- Research Institute, The Hospital for Sick Children, 686 Bay Street, Toronto M5G0A4, Canada
| | - Michael D. Wiese
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, 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 5001, Australia
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Research Institute, The Hospital for Sick Children, 686 Bay Street, Toronto M5G0A4, Canada
| |
Collapse
|
5
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [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: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
6
|
Dimasi CG, Darby JRT, Morrison JL. A change of heart: understanding the mechanisms regulating cardiac proliferation and metabolism before and after birth. J Physiol 2023; 601:1319-1341. [PMID: 36872609 PMCID: PMC10952280 DOI: 10.1113/jp284137] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/17/2023] [Indexed: 03/07/2023] Open
Abstract
Mammalian cardiomyocytes undergo major maturational changes in preparation for birth and postnatal life. Immature cardiomyocytes contribute to cardiac growth via proliferation and thus the heart has the capacity to regenerate. To prepare for postnatal life, structural and metabolic changes associated with increased cardiac output and function must occur. This includes exit from the cell cycle, hypertrophic growth, mitochondrial maturation and sarcomeric protein isoform switching. However, these changes come at a price: the loss of cardiac regenerative capacity such that damage to the heart in postnatal life is permanent. This is a significant barrier to the development of new treatments for cardiac repair and contributes to heart failure. The transitional period of cardiomyocyte growth is a complex and multifaceted event. In this review, we focus on studies that have investigated this critical transition period as well as novel factors that may regulate and drive this process. We also discuss the potential use of new biomarkers for the detection of myocardial infarction and, in the broader sense, cardiovascular disease.
Collapse
Affiliation(s)
- Catherine G. Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| |
Collapse
|
7
|
Cardiac Hypertrophy and Related Dysfunctions in Cushing Syndrome Patients-Literature Review. J Clin Med 2022; 11:jcm11237035. [PMID: 36498610 PMCID: PMC9739690 DOI: 10.3390/jcm11237035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The survival rate of adrenal Cushing syndrome patients has been greatly increased because of the availability of appropriate surgical and pharmacological treatments. Nevertheless, increased possibility of a heart attack induced by a cardiovascular event remains a major risk factor for the survival of affected patients. In experimental studies, hypercortisolemia has been found to cause cardiomyocyte hypertrophy via glucocorticoid receptor activation, including the possibility of cross talk among several hypertrophy signals related to cardiomyocytes and tissue-dependent regulation of 11β-hydroxysteroid dehydrogenase type 1. However, the factors are more complex in clinical cases, as both geometric and functional impairments leading to heart failure have been revealed, and their associations with a wide range of factors such as hypertension are crucial. In addition, knowledge regarding such alterations in autonomous cortisol secretion, which has a high risk of leading to heart attack as well as overt Cushing syndrome, is quite limited. When considering the effects of treatment, partial improvement of structural alterations is expected, while functional disorders are controversial. Therefore, whether the normalization of excess cortisol attenuates the risk related to cardiac hypertrophy has yet to be fully elucidated.
Collapse
|
8
|
Fowden AL, Vaughan OR, Murray AJ, Forhead AJ. Metabolic Consequences of Glucocorticoid Exposure before Birth. Nutrients 2022; 14:nu14112304. [PMID: 35684104 PMCID: PMC9182938 DOI: 10.3390/nu14112304] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids have an important role in development of the metabolic phenotype in utero. They act as environmental and maturational signals in adapting feto-placental metabolism to maximize the chances of survival both before and at birth. They influence placental nutrient handling and fetal metabolic processes to support fetal growth, fuel storage and energy production with respect to nutrient availability. More specifically, they regulate the transport, utilization and production of a range of nutrients by the feto-placental tissues that enables greater metabolic flexibility in utero while minimizing any further drain on maternal resources during periods of stress. Near term, the natural rise in fetal glucocorticoid concentrations also stimulates key metabolic adaptations that prepare tissues for the new energy demanding functions after birth. Glucocorticoids, therefore, have a central role in the metabolic communication between the mother, placenta and fetus that optimizes offspring metabolic phenotype for survival to reproductive age. This review discusses the effects of maternal and fetal glucocorticoids on the supply and utilization of nutrients by the feto-placental tissues with particular emphasis on studies using quantitative methods to assess metabolism in rodents and sheep in vivo during late pregnancy. It considers the routes of glucocorticoid overexposure in utero, including experimental administration of synthetic glucocorticoids, and the mechanisms by which these hormones control feto-placental metabolism at the molecular, cellular and systems levels. It also briefly examines the consequences of intrauterine glucocorticoid overexposure for postnatal metabolic health and the generational inheritance of metabolic phenotype.
Collapse
Affiliation(s)
- Abigail L. Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
- Correspondence:
| | - Owen R. Vaughan
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK;
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
| | - Alison J. Forhead
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| |
Collapse
|
9
|
Ghnenis A, Padmanabhan V, Vyas A. Sexual dimorphism in testosterone programming of cardiomyocyte development in sheep. Am J Physiol Heart Circ Physiol 2022; 322:H607-H621. [PMID: 35119334 PMCID: PMC8957338 DOI: 10.1152/ajpheart.00691.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
Perturbed in utero hormone milieu leads to intrauterine growth retardation (IUGR), a known risk factor for left ventricular (LV) dysfunction later in life. Gestational testosterone (T) excess predisposes offspring to IUGR and leads to LV myocardial disarray and hypertension in adult females. However, the early impact of T excess on LV programming and if it is female specific is unknown. LV tissues were obtained at day 90 gestation from days 30-90 T-treated or control fetuses (n = 6/group/sex) and morphometric and molecular analyses were conducted. Gestational T treatment increased cardiomyocyte number only in female fetuses. T excess upregulated receptor expression of insulin and insulin-like growth factor. Furthermore, in a sex-specific manner, T increased expression of phosphatidylinositol 3-kinase (PI3K) while downregulating phosphorylated mammalian target of rapamycin (pmTOR)-to-mTOR ratio suggestive of compensatory response. T excess 1) upregulated atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), markers of stress and cardiac hypertrophy and 2) upregulated estrogen receptors1 (ESR1) and 2 (ESR2), but not in androgen receptor (AR). Thus, gestational T excess upregulated markers of cardiac stress and hypertrophy in both sexes while inducing cardiomyocyte hyperplasia only in females, likely mediated via insulin and estrogenic programming.NEW & NOTEWORTHY The present study demonstrates sex-specific effects of gestational T excess between days 30 and 90 of gestation on the cardiac phenotype. Furthermore, the sex-specific programming is likely secondary to perturbation in both estrogen and insulin signaling pathways collectively. These findings are supportive of the role of androgen excess to serve as early biomarkers of CVD and could be critical in identifying therapeutic targets for LV hypertrophy and predict long-term CVD.
Collapse
Affiliation(s)
- Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | - Arpita Vyas
- College of Human Medicine, California Northstate University, Elk Grove, California
| |
Collapse
|
10
|
Bongiovanni C, Sacchi F, Da Pra S, Pantano E, Miano C, Morelli MB, D'Uva G. Reawakening the Intrinsic Cardiac Regenerative Potential: Molecular Strategies to Boost Dedifferentiation and Proliferation of Endogenous Cardiomyocytes. Front Cardiovasc Med 2021; 8:750604. [PMID: 34692797 PMCID: PMC8531484 DOI: 10.3389/fcvm.2021.750604] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Despite considerable efforts carried out to develop stem/progenitor cell-based technologies aiming at replacing and restoring the cardiac tissue following severe damages, thus far no strategies based on adult stem cell transplantation have been demonstrated to efficiently generate new cardiac muscle cells. Intriguingly, dedifferentiation, and proliferation of pre-existing cardiomyocytes and not stem cell differentiation represent the preponderant cellular mechanism by which lower vertebrates spontaneously regenerate the injured heart. Mammals can also regenerate their heart up to the early neonatal period, even in this case by activating the proliferation of endogenous cardiomyocytes. However, the mammalian cardiac regenerative potential is dramatically reduced soon after birth, when most cardiomyocytes exit from the cell cycle, undergo further maturation, and continue to grow in size. Although a slow rate of cardiomyocyte turnover has also been documented in adult mammals, both in mice and humans, this is not enough to sustain a robust regenerative process. Nevertheless, these remarkable findings opened the door to a branch of novel regenerative approaches aiming at reactivating the endogenous cardiac regenerative potential by triggering a partial dedifferentiation process and cell cycle re-entry in endogenous cardiomyocytes. Several adaptations from intrauterine to extrauterine life starting at birth and continuing in the immediate neonatal period concur to the loss of the mammalian cardiac regenerative ability. A wide range of systemic and microenvironmental factors or cell-intrinsic molecular players proved to regulate cardiomyocyte proliferation and their manipulation has been explored as a therapeutic strategy to boost cardiac function after injuries. We here review the scientific knowledge gained thus far in this novel and flourishing field of research, elucidating the key biological and molecular mechanisms whose modulation may represent a viable approach for regenerating the human damaged myocardium.
Collapse
Affiliation(s)
- Chiara Bongiovanni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Silvia Da Pra
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Elvira Pantano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Marco Bruno Morelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Gabriele D'Uva
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| |
Collapse
|
11
|
Effect of Preterm Birth on Cardiac and Cardiomyocyte Growth and the Consequences of Antenatal and Postnatal Glucocorticoid Treatment. J Clin Med 2021; 10:jcm10173896. [PMID: 34501343 PMCID: PMC8432182 DOI: 10.3390/jcm10173896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022] Open
Abstract
Preterm birth coincides with a key developmental window of cardiac growth and maturation, and thus has the potential to influence long-term cardiac function. Individuals born preterm have structural cardiac remodelling and altered cardiac growth and function by early adulthood. The evidence linking preterm birth and cardiovascular disease in later life is mounting. Advances in the perinatal care of preterm infants, such as glucocorticoid therapy, have improved survival rates, but at what cost? This review highlights the short-term and long-term impact of preterm birth on the structure and function of the heart and focuses on the impact of antenatal and postnatal glucocorticoid treatment on the immature preterm heart.
Collapse
|
12
|
Jellyman JK, Fletcher AJW, Fowden AL, Giussani DA. Glucocorticoid Maturation of Fetal Cardiovascular Function. Trends Mol Med 2020; 26:170-184. [PMID: 31718939 DOI: 10.1016/j.molmed.2019.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 12/27/2022]
Abstract
The last decade has seen rapid advances in the understanding of the central role of glucocorticoids in preparing the fetus for life after birth. However, relative to other organ systems, maturation by glucocorticoids of the fetal cardiovascular system has been ignored. Here, we review the effects of glucocorticoids on fetal basal cardiovascular function and on the fetal cardiovascular defense responses to acute stress. This is important because glucocorticoid-driven maturational changes in fetal cardiovascular function under basal and stressful conditions are central to the successful transition from intra- to extrauterine life. The cost-benefit balance for the cardiovascular health of the preterm baby of antenatal glucocorticoid therapy administered to pregnant women threatened with preterm birth is also discussed.
Collapse
Affiliation(s)
- Juanita K Jellyman
- Department of Biological Sciences, California State Polytechnic University, Pomona, CA, USA.
| | | | - Abigail L Fowden
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK; Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK; Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge, UK.
| |
Collapse
|
13
|
Bowen ME, Selzman CH, McKellar SH. Right Ventricular Involution: Big Changes in Small Hearts. J Surg Res 2019; 243:255-264. [PMID: 31252349 DOI: 10.1016/j.jss.2019.05.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Before birth, the fetal right ventricle (RV) is the pump for the systemic circulation and is about as thick as the left ventricle (LV). After birth, the RV becomes the pump for the lower pressure pulmonary circulation, and the RV chamber elongates without change in its wall thickness. We hypothesize that the fetal RV may be a model of compensated RV hypertrophy, and understanding this process may aid in discovering therapeutic strategies for RV failure. METHODS We performed a literature review and identified pertinent articles from 1980 to present. RESULTS The following topics were identified to be most pertinent in right ventricular involution: morphologic and histologic changes of the RV, cellular proliferation and terminal differentiation, the effect of stress on RV development, excitation contraction coupling and inotropic response change over time, and the amount of apoptosis through RV development. CONCLUSIONS The RV changes on multiple levels after its transition from systemic to pulmonary circulation. Although published literature has variable results due partly from differences between animal models, the literature shows a clear need for more research in the field.
Collapse
Affiliation(s)
- Megan E Bowen
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah.
| | - Craig H Selzman
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah
| | - Stephen H McKellar
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah
| |
Collapse
|
14
|
Antolic A, Li M, Richards EM, Curtis CW, Wood CE, Keller-Wood M. Mechanisms of in utero cortisol effects on the newborn heart revealed by transcriptomic modeling. Am J Physiol Regul Integr Comp Physiol 2019; 316:R323-R337. [PMID: 30624972 PMCID: PMC6483213 DOI: 10.1152/ajpregu.00322.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 12/20/2022]
Abstract
We have identified effects of elevated maternal cortisol (induced by maternal infusion 1 mg·kg-1·day-1) on fetal cardiac maturation and function using an ovine model. Whereas short-term exposure (115-130-day gestation) increased myocyte proliferation and Purkinje fiber apoptosis, infusions until birth caused bradycardia with increased incidence of arrhythmias at birth and increased perinatal death, despite normal fetal cortisol concentrations from 130 days to birth. Statistical modeling of the transcriptomic changes in hearts at 130 and 140 days suggested that maternal cortisol excess disrupts cardiac metabolism. In the current study, we modeled pathways in the left ventricle (LV) and interventricular septum (IVS) of newborn lambs after maternal cortisol infusion from 115 days to birth. In both LV and IVS the transcriptomic model indicated over-representation of cell cycle genes and suggested disruption of cell cycle progression. Pathways in the LV involved in cardiac architecture, including SMAD and bone morphogenetic protein ( BMP) were altered, and collagen deposition was increased. Pathways in IVS related to metabolism, calcium signaling, and the actin cytoskeleton were altered. Comparison of the effects of maternal cortisol excess to the effects of normal maturation from day 140 to birth revealed that only 20% of the genes changed in the LV were consistent with normal maturation, indicating that chronic elevation of maternal cortisol alters normal maturation of the fetal myocardium. These effects of maternal cortisol on the cardiac transcriptome, which may be secondary to metabolic effects, are consistent with cardiac remodeling and likely contribute to the adverse impact of maternal stress on perinatal cardiac function.
Collapse
Affiliation(s)
- Andrew Antolic
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| | - Mengchen Li
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Elaine M Richards
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Celia W Curtis
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| |
Collapse
|
15
|
Walejko JM, Antolic A, Koelmel JP, Garrett TJ, Edison AS, Keller-Wood M. Chronic maternal cortisol excess during late gestation leads to metabolic alterations in the newborn heart. Am J Physiol Endocrinol Metab 2019; 316:E546-E556. [PMID: 30620638 PMCID: PMC6459297 DOI: 10.1152/ajpendo.00386.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Our laboratory has previously shown in an ovine model of pregnancy that abnormal elevations in maternal cortisol during late gestation lead to increased fetal cardiac arrhythmias and mortality during peripartum. Furthermore, transcriptomic analysis of the fetal heart suggested alterations in TCA cycle intermediates and lipid metabolites in animals exposed to excess cortisol in utero. Therefore, we utilized a sheep model of pregnancy to determine how chronic increases in maternal cortisol alter maternal and fetal serum before birth and neonatal cardiac metabolites and lipids at term. Ewes were either infused with 1 mg·kg-1·day-1 of cortisol starting at gestational day 115 ( n = 9) or untreated ( n = 6). Serum was collected from the mother and fetus (gestational day 125), and hearts were collected following birth. Proton nuclear magnetic resonance (1H-NMR) spectroscopy was conducted to measure metabolic profiles of newborn heart specimens as well as fetal and maternal serum specimens. Mass spectrometry was conducted to measure lipid profiles of newborn heart specimens. We observed alterations in amino acid and TCA cycle metabolism as well as lipid and glycerophospholipid metabolism in newborn hearts after excess maternal cortisol in late gestation. In addition, we observed alterations in amino acid and TCA cycle metabolites in fetal but not in maternal serum during late gestation. These results suggest that fetal exposure to excess maternal cortisol alters placental and fetal metabolism before birth and limits normal cardiac metabolic maturation, which may contribute to increased risk of peripartum cardiac arrhythmias observed in these animals or later life cardiomyopathies.
Collapse
Affiliation(s)
- Jacquelyn M Walejko
- Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Andrew Antolic
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| | - Jeremy P Koelmel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida , Gainesville, Florida
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida , Gainesville, Florida
| | - Arthur S Edison
- Departments of Genetics and Biochemistry and Molecular Biology, Institute of Bioinformatics, and Complex Carbohydrate Research Center, University of Georgia , Athens, Georgia
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| |
Collapse
|
16
|
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
|
17
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
18
|
Camm EJ, Botting KJ, Sferruzzi-Perri AN. Near to One's Heart: The Intimate Relationship Between the Placenta and Fetal Heart. Front Physiol 2018; 9:629. [PMID: 29997513 PMCID: PMC6029139 DOI: 10.3389/fphys.2018.00629] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/09/2018] [Indexed: 01/19/2023] Open
Abstract
The development of the fetal heart is exquisitely controlled by a multitude of factors, ranging from humoral to mechanical forces. The gatekeeper regulating many of these factors is the placenta, an external fetal organ. As such, resistance within the placental vascular bed has a direct influence on the fetal circulation and therefore, the developing heart. In addition, the placenta serves as the interface between the mother and fetus, controlling substrate exchange and release of hormones into both circulations. The intricate relationship between the placenta and fetal heart is appreciated in instances of clinical placental pathology. Abnormal umbilical cord insertion is associated with congenital heart defects. Likewise, twin-to-twin transfusion syndrome, where monochorionic twins have unequal sharing of their placenta due to inter-twin vascular anastomoses, can result in cardiac remodeling and dysfunction in both fetuses. Moreover, epidemiological studies have suggested a link between placental phenotypic traits and increased risk of cardiovascular disease in adult life. To date, the mechanistic basis of the relationships between the placenta, fetal heart development and later risk of cardiac dysfunction have not been fully elucidated. However, studies using environmental exposures and gene manipulations in experimental animals are providing insights into the pathways involved. Likewise, surgical instrumentation of the maternal and fetal circulations in large animal species has enabled the manipulation of specific humoral and mechanical factors to investigate their roles in fetal cardiac development. This review will focus on such studies and what is known to date about the link between the placenta and heart development.
Collapse
Affiliation(s)
- Emily J Camm
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
19
|
Le B, Sutherland MR, Black MJ. Maladaptive structural remodelling of the heart following preterm birth. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
20
|
Lock MC, Botting KJ, Tellam RL, Brooks D, Morrison JL. Adverse Intrauterine Environment and Cardiac miRNA Expression. Int J Mol Sci 2017; 18:ijms18122628. [PMID: 29210999 PMCID: PMC5751231 DOI: 10.3390/ijms18122628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/10/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022] Open
Abstract
Placental insufficiency, high altitude pregnancies, maternal obesity/diabetes, maternal undernutrition and stress can result in a poor setting for growth of the developing fetus. These adverse intrauterine environments result in physiological changes to the developing heart that impact how the heart will function in postnatal life. The intrauterine environment plays a key role in the complex interplay between genes and the epigenetic mechanisms that regulate their expression. In this review we describe how an adverse intrauterine environment can influence the expression of miRNAs (a sub-set of non-coding RNAs) and how these changes may impact heart development. Potential consequences of altered miRNA expression in the fetal heart include; Hypoxia inducible factor (HIF) activation, dysregulation of angiogenesis, mitochondrial abnormalities and altered glucose and fatty acid transport/metabolism. It is important to understand how miRNAs are altered in these adverse environments to identify key pathways that can be targeted using miRNA mimics or inhibitors to condition an improved developmental response.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Ross L Tellam
- Early Origins of Adult Health Research Group; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
- CSIRO Agriculture, 306 Carmody Rd, St. Lucia, QLD 4067, Australia.
| | - Doug Brooks
- Mechanisms in Cell Biology and Disease Research Group School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Janna L Morrison
- Early Origins of Adult Health Research Group; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
21
|
Antolic A, Wood CE, Keller-Wood M. Chronic maternal hypercortisolemia in late gestation alters fetal cardiac function at birth. Am J Physiol Regul Integr Comp Physiol 2017; 314:R342-R352. [PMID: 29092858 DOI: 10.1152/ajpregu.00296.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Studies in our laboratory have shown that modest chronic increases in maternal cortisol concentrations over the last 0.20 of gestation impair maternal glucose metabolism and increase the incidence of perinatal stillbirth. Previous studies had found that an increase in maternal cortisol concentrations from 115 to 130 days of gestation in sheep increased both proliferation in fetal cardiomyocytes and apoptosis in the fetal cardiac Purkinje fibers. We hypothesized that the adverse effects of excess cortisol may result in defects in cardiac conduction during labor and delivery. In the present study, we infused cortisol (1 mg·kg-1·day-1) into late gestation pregnant ewes and continuously monitored fetal aortic pressure and ECG through labor and delivery. We found that, although the fetuses of cortisol infused ewes had normal late gestation patterns of arterial pressure and heart rate, there was a significant decrease in fetal aortic pressure and heart rate on the day of birth, specifically in the final hour before delivery. Significant changes in the fetal ECG were also apparent on the day of birth, including prolongation of the P wave and P-R interval. We speculate that chronic exposure to glucocorticoids alters cardiac metabolism or ion homeostasis, contributing to cardiac dysfunction, precipitated by active labor and delivery.
Collapse
Affiliation(s)
- Andrew Antolic
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida , Gainesville, Florida
| |
Collapse
|
22
|
Johansen IB, Sandblom E, Skov PV, Gräns A, Ekström A, Lunde IG, Vindas MA, Zhang L, Höglund E, Frisk M, Sjaastad I, Nilsson GE, Øverli Ø. Bigger is not better: cortisol-induced cardiac growth and dysfunction in salmonids. ACTA ACUST UNITED AC 2017; 220:2545-2553. [PMID: 28476893 DOI: 10.1242/jeb.135046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/02/2017] [Indexed: 01/04/2023]
Abstract
Stress and elevated cortisol levels are associated with pathological heart growth and cardiovascular disease in humans and other mammals. We recently established a link between heritable variation in post-stress cortisol production and cardiac growth in salmonid fish too. A conserved stimulatory effect of the otherwise catabolic steroid hormone cortisol is probably implied, but has to date not been established experimentally. Furthermore, whereas cardiac growth is associated with failure of the mammalian heart, pathological cardiac hypertrophy has not previously been described in fish. Here, we show that rainbow trout (Oncorhynchus mykiss) treated with cortisol in the diet for 45 days have enlarged hearts with lower maximum stroke volume and cardiac output. In accordance with impaired cardiac performance, overall circulatory oxygen-transporting capacity was diminished as indicated by reduced aerobic swimming performance. In contrast to the well-known adaptive/physiological heart growth observed in fish, cortisol-induced growth is maladaptive. Furthermore, the observed heart growth was associated with up-regulated signature genes of mammalian cardiac pathology, suggesting that signalling pathways mediating cortisol-induced cardiac remodelling in fish are conserved from fish to mammals. Altogether, we show that excessive cortisol can induce pathological cardiac remodelling. This is the first study to report and integrate the etiology, physiology and molecular biology of cortisol-induced pathological remodelling in fish.
Collapse
Affiliation(s)
- Ida B Johansen
- Department of Biosciences, University of Oslo, Blindern, 0316 Oslo, Norway .,Bjørknes College, 0456 Oslo, Norway.,Department of Food Safety and Infection Biology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Erik Sandblom
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Peter V Skov
- National Institute of Aquatic Resources, Technical University of Denmark, North Sea Science Centre, Section for Aquaculture, 9850 Hirtshals, Denmark
| | - Albin Gräns
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, 532 23 Skara, Sweden
| | - Andreas Ekström
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway.,KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0424 Oslo, Norway.,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Marco A Vindas
- Department of Biosciences, University of Oslo, Blindern, 0316 Oslo, Norway
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway.,KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0424 Oslo, Norway
| | - Erik Höglund
- Norwegian Institute of Water Research (NIVA), Gaustadalléen 21, 0349 Oslo, Norway.,Centre of Coastal Research, University of Agder, PO Box 422, 4604 Kristiansand, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway.,KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0424 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway.,KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0424 Oslo, Norway
| | - Göran E Nilsson
- Department of Biosciences, University of Oslo, Blindern, 0316 Oslo, Norway
| | - Øyvind Øverli
- Department of Food Safety and Infection Biology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| |
Collapse
|
23
|
Wang KCW, Botting KJ, Zhang S, McMillen IC, Brooks DA, Morrison JL. Akt signaling as a mediator of cardiac adaptation to low birth weight. J Endocrinol 2017; 233:R81-R94. [PMID: 28219933 DOI: 10.1530/joe-17-0039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2022]
Abstract
Intrauterine insults, such as poor nutrition and placental insufficiency, can alter cardiomyocyte development, and this can have significant long-term implications for heart health. Consequently, epidemiological studies have shown that low-birth-weight babies have an increased risk of death from cardiovascular disease in adult life. In addition, intrauterine growth restriction can result in increased left ventricular hypertrophy, which is the strongest predictor for poor health outcomes in cardiac patients. The mechanisms responsible for these associations are not clear, but a suboptimal intrauterine environment can program alternative expression of genes such as cardiac IGF-2/H19, IGF-2R and AT1R through either an increase or decrease in DNA methylation or histone acetylation at specific loci. Furthermore, hypoxia and other intrauterine insults can also activate the IGF-1 receptor via IGF-1 and IGF-2, and the AT1 receptor via angiotensin signaling pathways; both of which can result in the phosphorylation of Akt and the activation of a range of downstream pathways. In turn, Akt activation can increase cardiac angiogenesis and cardiomyocyte apoptosis and promote a reversion of metabolism in postnatal life to a fetal phenotype, which involves increased reliance on glucose. Cardiac Akt can also be indirectly regulated by microRNAs and conversely can target microRNAs that will eventually affect other specific cardiac genes and proteins. This review aims to discuss our understanding of this complex network of interactions, which may help explain the link between low birth weight and the increased risk of cardiovascular disease in adult life.
Collapse
Affiliation(s)
- Kimberley C W Wang
- Early Origins of Adult Health Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Kimberley J Botting
- Early Origins of Adult Health Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Song Zhang
- Early Origins of Adult Health Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research GroupSchool of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Gay MS, Dasgupta C, Li Y, Kanna A, Zhang L. Dexamethasone Induces Cardiomyocyte Terminal Differentiation via Epigenetic Repression of Cyclin D2 Gene. J Pharmacol Exp Ther 2016; 358:190-8. [PMID: 27302109 PMCID: PMC4959091 DOI: 10.1124/jpet.116.234104] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022] Open
Abstract
Dexamethasone treatment of newborn rats inhibited cardiomyocyte proliferation and stimulated premature terminal differentiation of cardiomyocytes in the developing heart. Yet mechanisms remain undetermined. The present study tested the hypothesis that the direct effect of glucocorticoid receptor-mediated epigenetic repression of cyclin D2 gene in the cardiomyocyte plays a key role in the dexamethasone-mediated effects in the developing heart. Cardiomyocytes were isolated from 2-day-old rats. Cells were stained with a cardiomyocyte marker α-actinin and a proliferation marker Ki67. Cyclin D2 expression was evaluated by Western blot and quantitative real-time polymerase chain reaction. Promoter methylation of CcnD2 was determined by methylated DNA immunoprecipitation (MeDIP). Overexpression of Cyclin D2 was conducted by transfection of FlexiCcnD2 (+CcnD2) construct. Treatment of cardiomyocytes isolated from newborn rats with dexamethasone for 48 hours significantly inhibited cardiomyocyte proliferation with increased binucleation and decreased cyclin D2 protein abundance. These effects were blocked with Ru486 (mifepristone). In addition, the dexamethasone treatment significantly increased cyclin D2 gene promoter methylation in newborn rat cardiomyocytes. 5-Aza-2'-deoxycytidine inhibited dexamethasone-mediated promoter methylation, recovered dexamethasone-induced cyclin D2 gene repression, and blocked the dexamethasone-elicited effects on cardiomyocyte proliferation and binucleation. In addition, the overexpression of cyclin D2 restored the dexamethasone-mediated inhibition of proliferation and increase in binucleation in newborn rat cardiomyocytes. The results demonstrate that dexamethasone acting on glucocorticoid receptors has a direct effect and inhibits proliferation and stimulates premature terminal differentiation of cardiomyocytes in the developing heart via epigenetic repression of cyclin D2 gene.
Collapse
Affiliation(s)
- Maresha S Gay
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Chiranjib Dasgupta
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Yong Li
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Angela Kanna
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
25
|
Schubert U, Müller M, Abdul-Khaliq H, Norman M. Preterm Birth Is Associated with Altered Myocardial Function in Infancy. J Am Soc Echocardiogr 2016; 29:670-8. [PMID: 27156903 DOI: 10.1016/j.echo.2016.03.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Preterm birth has been associated with myocardial remodeling and accelerated cardiovascular ageing in later life, but the underlying mechanisms are unknown. The investigators used echocardiography to undertake a sequential analysis of myocardial function in preterm infants. METHODS This study evaluated the cardiac performance of 25 very preterm infants (born at a gestational age of 26-30 weeks), at birth, 3 months (term-equivalent age), and 6 months later (3 months of corrected age). Speckle-tracking echocardiography was used to determine myocardial function, assessing the magnitude of myocardial deformation as longitudinal strain, deformation rate (strain rate), and velocity in both ventricles during systole and diastole. The results were compared with those in 30 infants born at term investigated at birth and at 3 months of age. RESULTS At term-equivalent age, the speckle-tracking estimates were similar in both groups. Three months later, very preterm infants exhibited significantly lower left ventricular mean free wall longitudinal strain (-20.0% vs -22.0%, P = .010) and lower left ventricular early diastolic (median, -7.37 vs -10.9 cm/sec, P = .003) and late diastolic (median, -5.11 vs -6.95 cm/sec, P = .009) myocardial velocities than infants born at term. There were no statistically significant group differences in right ventricular or interventricular septal measurements. Conventional echocardiographic variables did not differ significantly between the two groups at any age. CONCLUSIONS Very preterm infants develop altered left ventricular myocardial function 6 months after birth. Follow-up examinations are needed to determine the implications for cardiovascular health in the growing number of children surviving very preterm birth.
Collapse
Affiliation(s)
- Ulf Schubert
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Matthias Müller
- Department of Pediatric Cardiology, University Hospital Homburg (Saar), Homburg, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, University Hospital Homburg (Saar), Homburg, Germany
| | - Mikael Norman
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Bensley JG, De Matteo R, Harding R, Black MJ. The effects of preterm birth and its antecedents on the cardiovascular system. Acta Obstet Gynecol Scand 2016; 95:652-63. [PMID: 26918772 DOI: 10.1111/aogs.12880] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/09/2016] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Preterm birth occurs in approximately 10% of all births worldwide. It prematurely exposes the developing cardiovascular system to the hemodynamic transition that occurs at birth and to the subsequent functional demands of life ex utero. This review describes the current knowledge of the effects of preterm birth, and some of its common antecedents (chorioamnionitis, intra-uterine growth restriction, and maternal antenatal corticosteroid administration), on the structure of the myocardium. MATERIAL AND METHODS A thorough literature search was conducted for articles relating to how preterm birth, and its antecedents, affect development of the heart. Given that sheep are an excellent model for the studies of cardiac development, this review has focused on experimental studies in sheep as well as clinical findings. RESULTS Our review of the literature demonstrates that individuals born preterm are at an increased risk of cardiovascular disease later in life, including increased mean arterial pressure, abnormally shaped and sub-optimally performing hearts and changes in the vasculature. The review highlights how antenatal corticosteroids, intra-uterine growth restriction, and exposure to chorioamnionitis also have the potential to impact cardiac growth in the preterm newborn. CONCLUSIONS Preterm birth and its common antecedents (antenatal corticosteroids, intra-uterine growth restriction, and chorioamnionitis) have the potential to adversely impact cardiac structure immediately following birth and in later life.
Collapse
Affiliation(s)
- Jonathan G Bensley
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Mary J Black
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Jonker SS, Louey S. Endocrine and other physiologic modulators of perinatal cardiomyocyte endowment. J Endocrinol 2016; 228:R1-18. [PMID: 26432905 PMCID: PMC4677998 DOI: 10.1530/joe-15-0309] [Citation(s) in RCA: 43] [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] [Accepted: 10/01/2015] [Indexed: 01/09/2023]
Abstract
Immature contractile cardiomyocytes proliferate to rapidly increase cell number, establishing cardiomyocyte endowment in the perinatal period. Developmental changes in cellular maturation, size and attrition further contribute to cardiac anatomy. These physiological processes occur concomitant with a changing hormonal environment as the fetus prepares itself for the transition to extrauterine life. There are complex interactions between endocrine, hemodynamic and nutritional regulators of cardiac development. Birth has been long assumed to be the trigger for major differences between the fetal and postnatal cardiomyocyte growth patterns, but investigations in normally growing sheep and rodents suggest this may not be entirely true; in sheep, these differences are initiated before birth, while in rodents they occur after birth. The aim of this review is to draw together our understanding of the temporal regulation of these signals and cardiomyocyte responses relative to birth. Further, we consider how these dynamics are altered in stressed and suboptimal intrauterine environments.
Collapse
Affiliation(s)
- S S Jonker
- Knight Cardiovascular Institute Center for Developmental HealthOregon Health and Science University, Portland, Oregon 97239, USA
| | - S Louey
- Knight Cardiovascular Institute Center for Developmental HealthOregon Health and Science University, Portland, Oregon 97239, USA
| |
Collapse
|
28
|
Antolic A, Feng X, Wood CE, Richards EM, Keller-Wood M. Increased maternal nighttime cortisol concentrations in late gestation alter glucose and insulin in the neonatal lamb. Physiol Rep 2015; 3:3/9/e12548. [PMID: 26371232 PMCID: PMC4600389 DOI: 10.14814/phy2.12548] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Previous studies in our laboratory have shown that a modest chronic increase in maternal cortisol concentrations impairs maternal glucose metabolism and increases the incidence of perinatal stillbirth. The dramatic outcomes prevented our ability to study the effects of maternal hypercortisolemia on neonatal growth, glucose metabolism, and hypothalamo–pituitary–adrenal axis response. Therefore, we developed a model in which pregnant ewes are infused for 12 h/day at 0.5 mg·kg–1·day–1 from day 115 of gestation until delivery (˜145), elevating nighttime plasma cortisol concentrations. This pattern of elevation of cortisol mimics that in patients with elevated evening cortisol concentrations, as in Cushing’s syndrome or chronic depression. Plasma cortisol, glucose, insulin, and electrolytes were measured during pregnancy and postpartum in control and cortisol-infused ewes and their postnatal lambs for the first 14 days after delivery. Neonatal growth and plasma ACTH, aldosterone, renin activity, and electrolytes, and organ weights at 14 days of age were also measured. Infusion of cortisol increased maternal plasma cortisol during pregnancy but not postpartum, and did not alter neonatal ACTH or cortisol. Although maternal glucose and insulin concentrations were not changed by the maternal infusion of cortisol, neonatal plasma glucose was increased and plasma insulin was decreased compared to those in the control group. Neonatal ponderal index and kidney weight were reduced, left ventricular wall thickness was increased, and plasma sodium and creatinine were increased after maternal cortisol infusion. These results suggest that excess maternal cortisol concentrations in late gestation alter growth, glucose and insulin regulation, and organ maturation in the neonate.
Collapse
Affiliation(s)
- Andrew Antolic
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Xiaodi Feng
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Elaine M Richards
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | | |
Collapse
|
29
|
Forhead AJ, Jellyman JK, De Blasio MJ, Johnson E, Giussani DA, Broughton Pipkin F, Fowden AL. Maternal Dexamethasone Treatment Alters Tissue and Circulating Components of the Renin-Angiotensin System in the Pregnant Ewe and Fetus. Endocrinology 2015; 156:3038-46. [PMID: 26039155 PMCID: PMC4511127 DOI: 10.1210/en.2015-1197] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antenatal synthetic glucocorticoids promote fetal maturation in pregnant women at risk of preterm delivery and their mechanism of action may involve other endocrine systems. This study investigated the effect of maternal dexamethasone treatment, at clinically relevant doses, on components of the renin-angiotensin system (RAS) in the pregnant ewe and fetus. From 125 days of gestation (term, 145 ± 2 d), 10 ewes carrying single fetuses of mixed sex (3 female, 7 male) were injected twice im, at 10-11 pm, with dexamethasone (2 × 12 mg, n = 5) or saline (n = 5) at 24-hour intervals. At 10 hours after the second injection, maternal dexamethasone treatment increased angiotensin-converting enzyme (ACE) mRNA levels in the fetal lungs, kidneys, and heart and ACE concentration in the circulation and lungs, but not kidneys, of the fetuses. Fetal cardiac mRNA abundance of angiotensin II (AII) type 2 receptor decreased after maternal dexamethasone treatment. Between the two groups of fetuses, there were no significant differences in plasma angiotensinogen or renin concentrations; in transcript levels of renal renin, or AII type 1 or 2 receptors in the lungs and kidneys; or in pulmonary, renal or cardiac protein content of the AII receptors. In the pregnant ewes, dexamethasone administration increased pulmonary ACE and plasma angiotensinogen, and decreased plasma renin, concentrations. Some of the effects of dexamethasone treatment on the maternal and fetal RAS were associated with altered insulin and thyroid hormone activity. Changes in the local and circulating RAS induced by dexamethasone exposure in utero may contribute to the maturational and tissue-specific actions of antenatal glucocorticoid treatment.
Collapse
Affiliation(s)
- Alison J Forhead
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Juanita K Jellyman
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Miles J De Blasio
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Emma Johnson
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Fiona Broughton Pipkin
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| | - Abigail L Fowden
- Department of Physiology, Development and Neuroscience (A.J.F., J.K.J., M.J.D.B., E.J., D.A.G., A.L.F.), University of Cambridge, Cambridge CB2 3EG, United Kingdom; Department of Biological and Medical Sciences (A.J.F.), Oxford Brookes University, Oxford OX3 0BP, United Kingdom; and Department of Obstetrics and Gynaecology (F.B.P.), University of Nottingham, Nottingham NG5 1PB, United Kingdom
| |
Collapse
|
30
|
Richards EM, Wood CE, Rabaglino MB, Antolic A, Keller-Wood M. Mechanisms for the adverse effects of late gestational increases in maternal cortisol on the heart revealed by transcriptomic analyses of the fetal septum. Physiol Genomics 2014; 46:547-59. [PMID: 24867915 DOI: 10.1152/physiolgenomics.00009.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We have previously shown in sheep that 10 days of modest chronic increase in maternal cortisol resulting from maternal infusion of cortisol (1 mg/kg/day) caused fetal heart enlargement and Purkinje cell apoptosis. In subsequent studies we extended the cortisol infusion to term, finding a dramatic incidence of stillbirth in the pregnancies with chronically increased cortisol. To investigate effects of maternal cortisol on the heart, we performed transcriptomic analyses on the septa using ovine microarrays and Webgestalt and Cytoscape programs for pathway inference. Analyses of the transcriptomic effects of maternal cortisol infusion for 10 days (130 day cortisol vs 130 day control), or ∼25 days (140 day cortisol vs 140 day control) and of normal maturation (140 day control vs 130 day control) were performed. Gene ontology terms related to immune function and cytokine actions were significantly overrepresented as genes altered by both cortisol and maturation in the septa. After 10 days of cortisol, growth factor and muscle cell apoptosis pathways were significantly overrepresented, consistent with our previous histologic findings. In the term fetuses (∼25 days of cortisol) nutrient pathways were significantly overrepresented, consistent with altered metabolism and reduced mitochondria. Analysis of mitochondrial number by mitochondrial DNA expression confirmed a significant decrease in mitochondria. The metabolic pathways modeled as altered by cortisol treatment to term were different from those modeled during maturation of the heart to term, and thus changes in gene expression in these metabolic pathways may be indicative of the fetal heart pathophysiologies seen in pregnancies complicated by stillbirth, including gestational diabetes, Cushing's disease and chronic stress.
Collapse
Affiliation(s)
- Elaine M Richards
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Maria Belen Rabaglino
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Andrew Antolic
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| |
Collapse
|
31
|
Kim MY, Eiby YA, Lumbers ER, Wright LL, Gibson KJ, Barnett AC, Lingwood BE. Effects of glucocorticoid exposure on growth and structural maturation of the heart of the preterm piglet. PLoS One 2014; 9:e93407. [PMID: 24676209 PMCID: PMC3968162 DOI: 10.1371/journal.pone.0093407] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 03/05/2014] [Indexed: 11/27/2022] Open
Abstract
Inadequate maintenance of systemic blood flow in neonates following preterm birth is associated with increased morbidity and mortality, and may be due in part to structural immaturity of the myocardium. Maternal glucocorticoid administration is associated with improved cardiovascular function, and possibly promotes structural maturation of the myocardium. This study assessed the structural maturity of the myocardium in male and female preterm and term piglets, and preterm piglets exposed to a regimen of maternal glucocorticoids as used clinically. In preterm, term and glucocorticoid exposed preterm piglets cardiomyocyte maturity was examined by measuring the proportion of binucleated myocytes and the volumes of single living ventricular cardiomyocytes with fluorescence microscopy. Ventricular apoptosis and proliferation were measured by immunohistochemistry. Preterm piglet hearts had fewer binucleated myocytes, smaller myocytes, and more proliferative and fewer apoptotic nuclei than term hearts. Maternal glucocorticoid treatment resulted in increased binucleation with no increase in myocyte volume, and levels of proliferation and apoptosis that were more similar to the term heart. Atrial weights were increased and in female piglets there was an increase in the ratio of left to right ventricular weight. The observed changes in atrial mass and myocyte structural maturation correlated with changes in cardiac function of isolated hearts of littermates. In conclusion, the association between increased myocardial maturation following glucocorticoid exposure, improved cardiac function in littermates, and clinical improvement in human neonatal cardiac function exposed to antenatal glucocorticoids, suggests that glucocorticoid exposure contributes to improved cardiovascular function in preterm infants by promoting myocardial structural maturity.
Collapse
Affiliation(s)
- Min Young Kim
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Yvonne A. Eiby
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Eugenie R. Lumbers
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Layne L. Wright
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Karen J. Gibson
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Amanda C. Barnett
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Barbara E. Lingwood
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
- * E-mail:
| |
Collapse
|
32
|
Kim MY, Finch AM, Lumbers ER, Boyce AC, Gibson KJ, Eiby YA, Lingwood BE. Expression of adrenoceptor subtypes in preterm piglet heart is different to term heart. PLoS One 2014; 9:e92167. [PMID: 24670668 PMCID: PMC3966759 DOI: 10.1371/journal.pone.0092167] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 02/19/2014] [Indexed: 11/18/2022] Open
Abstract
Preterm delivery increases the risk of inadequate systemic blood flow and hypotension, and many preterm infants fail to respond to conventional inotrope treatments. If the profile of cardiac adrenoceptor subtypes in the preterm neonate is different to that at term this may contribute to these clinical problems. This study measured mRNA expression of β1, β2, α1A, α2A and α2B-adrenoceptor subtypes by real time PCR in term (113d), preterm (91d) and preterm piglets (91d) exposed to maternal glucocorticoid treatment. Abundance of β-adrenoceptor binding sites in the left ventricle was measured using saturation binding assays. Relative abundance of β1-adrenoceptor mRNA in untreated preterm hearts was ∼50% of term abundance in both left and right ventricles (P<0.001). Trends in receptor binding site density measurements supported this observation (P = 0.07). Glucocorticoid exposure increased β1-adrenoceptor mRNA levels in the right ventricle of preterm hearts (P = 0.008) but did not alter expression in the left ventricle (P>0.1). Relative abundance of α1A-adrenoceptor mRNA was the same in preterm and term piglet hearts (P = >0.1) but was reduced by maternal glucocorticoid treatment (P<0.01); α2A-adrenoceptor mRNA abundance was higher in untreated and glucocorticoid exposed preterm piglet hearts than in term piglets (P<0.001). There was no difference between male and female piglets in mRNA abundance of any of the genes studied. In conclusion, there is reduced mRNA abundance of β1-adrenoceptors in the preterm pig heart. If this lower expression of β-adrenoceptors occurs in human preterm infants, it could explain their poor cardiovascular function and their frequent failure to respond to commonly used inotropes.
Collapse
MESH Headings
- Animals
- Binding Sites
- Female
- Gene Expression Regulation, Developmental
- Heart/embryology
- Male
- Myocardium/metabolism
- Premature Birth/metabolism
- Receptors, Adrenergic/genetics
- Receptors, Adrenergic/metabolism
- Receptors, Adrenergic, alpha/genetics
- Receptors, Adrenergic, alpha/metabolism
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta/metabolism
- Sus scrofa/embryology
- Term Birth/metabolism
Collapse
Affiliation(s)
- Min Young Kim
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Angela M. Finch
- Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Eugenie R. Lumbers
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
- Department of Physiology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Amanda C. Boyce
- Department of Physiology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Karen J. Gibson
- Department of Physiology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Yvonne A. Eiby
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | - Barbara E. Lingwood
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
- * E-mail:
| |
Collapse
|
33
|
Feng X, Reini SA, Richards E, Wood CE, Keller-Wood M. Cortisol stimulates proliferation and apoptosis in the late gestation fetal heart: differential effects of mineralocorticoid and glucocorticoid receptors. Am J Physiol Regul Integr Comp Physiol 2013; 305:R343-50. [PMID: 23785077 DOI: 10.1152/ajpregu.00112.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We have previously found that modest chronic increases in maternal cortisol result in an enlarged fetal heart. To explore the mechanisms of this effect, we used intrapericardial infusions of a mineralocorticoid receptor (MR) antagonist (canrenoate) or of a glucocorticoid receptor (GR) antagonist (mifepristone) in the fetus during maternal infusion of cortisol (1 mg·kg⁻¹·day⁻¹). We have shown that the MR antagonist blocked the increase in fetal heart weight and in wall thickness resulting from maternal cortisol infusion. In the current study we extended those studies and found that cortisol increased Ki67 staining in both ventricles, indicating cell proliferation, but also increased active caspase-3 staining in cells of the conduction pathway in the septum and subendocardial layers of the left ventricle, suggesting increased apoptosis in Purkinje fibers. The MR antagonist blocked the increase in cell proliferation, whereas the GR antagonist blocked the increased apoptosis in Purkinje fibers. We also found evidence of activation of caspase-3 in c-kit-positive cells, suggesting apoptosis in stem cell populations in the ventricle. These studies suggest a potentially important role of corticosteroids in the terminal remodeling of the late gestation fetal heart and suggest a mechanism for the cardiac enlargement with excess corticosteroid exposure.
Collapse
Affiliation(s)
- Xiaodi Feng
- Departments of Pharmacodynamics, and Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | | | | | | | | |
Collapse
|
34
|
Nesan D, Vijayan MM. Role of glucocorticoid in developmental programming: evidence from zebrafish. Gen Comp Endocrinol 2013; 181:35-44. [PMID: 23103788 DOI: 10.1016/j.ygcen.2012.10.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 10/13/2012] [Indexed: 10/27/2022]
Abstract
The vertebrate corticosteroid stress response is highly conserved and a key function is to restore homeostasis by mobilizing and reallocating energy stores. This process is primarily initiated by activation of the hypothalamus-pituitary-adrenal axis, leading to the release of corticosteroids into the circulation. In teleosts, cortisol is the primary corticosteroid that is released into the circulation in response to stress. This steroid activates corticosteroid receptors that are ligand-bound transcription factors, modulating downstream gene expression in target tissues. Recent research in zebrafish (Danio rerio) has identified novel roles for cortisol in early developmental processes, including organogenesis and mesoderm formation. As cortisol biosynthesis commences only around the time of hatch in teleosts, the early developmental events are orchestrated by cortisol that is maternally deposited prior to fertilization. This review will highlight the molecular events leading to the development of the corticosteroid stress axis, and the possible role of cortisol in the developmental programming of stress axis function. Use of zebrafish as a model may lead to significant insights into the conserved role of glucocorticoids during early development with potential implications in biomedical research, including fetal stress syndromes in humans.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | |
Collapse
|
35
|
Nesan D, Vijayan MM. Embryo exposure to elevated cortisol level leads to cardiac performance dysfunction in zebrafish. Mol Cell Endocrinol 2012; 363:85-91. [PMID: 22842336 DOI: 10.1016/j.mce.2012.07.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 06/19/2012] [Accepted: 07/19/2012] [Indexed: 11/17/2022]
Abstract
In zebrafish (Danio rerio), de novo cortisol synthesis commences only after hatching, providing an interesting model to study the effects of maternal stress and abnormal cortisol deposition on embryo development and performance. We hypothesized that elevated cortisol levels during pre-hatch embryogenesis compromise cardiac performance in developing zebrafish. Cortisol was microinjected into one-cell embryos to elevate basal cortisol levels during embryogenesis. Elevated embryo cortisol content increased heart deformities, including pericardial edema and malformed chambers, and lowered resting heartbeat post-hatch. This phenotype coincided with suppression of key cardiac genes, including nkx2.5, cardiac myosin light chain 1, cardiac troponin type T2A, and calcium transporting ATPase, underpinning a mechanistic link to heart malformation. The attenuation of the heartbeat response to a secondary stressor post-hatch also confirms a functional reduction in cardiac performance. Altogether, high cortisol content during embryogenesis, mimicking increased deposition due to maternal stress, decreases cardiac performance and may reduce zebrafish offspring survival.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | |
Collapse
|
36
|
Wang KCW, Botting KJ, Padhee M, Zhang S, McMillen IC, Suter CM, Brooks DA, Morrison JL. Early origins of heart disease: Low birth weight and the role of the insulin-like growth factor system in cardiac hypertrophy. Clin Exp Pharmacol Physiol 2012; 39:958-64. [DOI: 10.1111/j.1440-1681.2012.05743.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Kimberley CW Wang
- Early Origins of Adult Health Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| | | | - Monalisa Padhee
- Early Origins of Adult Health Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| | - Song Zhang
- Early Origins of Adult Health Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| | - Catherine M Suter
- Victor Chang Cardiac Research Institute; Darlinghurst; New South Wales; Australia
| | - Doug A Brooks
- Cell Biology of Diseases Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group; Sansom Institute for Health Research; School of Pharmacy and Medical Sciences; University of Adelaide; Adelaide; South Australia; Australia
| |
Collapse
|
37
|
Eiby YA, Lumbers ER, Headrick JP, Lingwood BE. Left ventricular output and aortic blood flow in response to changes in preload and afterload in the preterm piglet heart. Am J Physiol Regul Integr Comp Physiol 2012; 303:R769-77. [PMID: 22895742 DOI: 10.1152/ajpregu.00010.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low systemic blood flow occurs in up to 30% of infants born at less than 30 wk gestation. It is associated with increased morbidity and mortality, and current treatments are ineffective in 40% of cases. The aim of this study was to assess the ability of the preterm heart to respond to the acute shifts in preload and afterload that occur at the time of birth. Myocardial and coronary vascular function was assessed using an isolated working heart model in term (115 days) and preterm (92 days) piglets. Cardiac output/kg body wt in preterm hearts was ∼50% lower than that of term hearts (P = 0.001). Pressure development was similar in term and preterm hearts. Elevations in preload increased cardiac output and aortic flow similarly in term and preterm hearts, demonstrating significant preload "reserve". By contrast, elevations in afterload markedly depressed aortic flow, with a greater proportion of cardiac output being distributed to coronary flow in preterm hearts at high afterloads. The demands of increased workload were associated with greater increases in coronary flow in preterm hearts compared with term hearts. In preterm hearts, exposure to maternal glucocorticoids resulted in increased aortic flow when afterload was below 25 mmHg. These data suggest the preterm heart lacks the functional capacity to acutely adapt to postnatal afterload. To maximize systemic blood flow in preterm infants, treatments limiting afterload, while harnessing significant preload reserve, should be further explored.
Collapse
Affiliation(s)
- Yvonne A Eiby
- The University of Queensland, Centre for Clinical Research, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
38
|
Social defeat and isolation induce clear signs of a depression-like state, but modest cardiac alterations in wild-type rats. Physiol Behav 2012; 106:142-50. [DOI: 10.1016/j.physbeh.2012.01.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/18/2012] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
|
39
|
Jiang P, Dai W, Yan S, Chen Z, Xu R, Ding J, Xiang L, Wang S, Liu R, Zhang W. Biomarkers in the early period of acute myocardial infarction in rat serum and protective effects of Shexiang Baoxin Pill using a metabolomic method. JOURNAL OF ETHNOPHARMACOLOGY 2011; 138:530-536. [PMID: 22001859 DOI: 10.1016/j.jep.2011.09.049] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 09/12/2011] [Accepted: 09/18/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE To identify the biomarkers in early period of acute myocardial infarction (AMI) in rat serum and reveal the effective mechanism of a Traditional Chinese Medicine (TCM) named Shexiang Baoxin Pill (SBP). MATERIAL AND METHOD A metabolomic approach using reversed-phase liquid chromatography/quadrupole time-of-flight mass spectrometry (LC-Q-TOF-MS) was developed. RESULTS Fourteen biomarkers in the early period of acute myocardial infarction (AMI) in rat serum were identified. These biomarkers include 5-methylcytosine, cystathionine ketimine, 2-oxoadipic acid, thymidine, epinephrine, homocystine, uric acid, 12(S)-hydroperoxyeicosatetraenoic acid (12s-HPETE), 11-dehydrocorticosterone, 12(S)-hydroxyeicosatetraenoic acid (12s-HETE), deoxycorticosterone, corticosterone, aldosterone and cortisol. Through pathway analysis of these biomarkers, inflammation, hypertrophy and oxidative injury were considered the most relevant pathological changes in early period of AMI. CONCLUSION Identification of AMI biomarkers not only supplied a systematic view of the progression of AMI in the early period but also provided the theoretical basis for the prevention or treatment of AMI. The results demonstrated that SBP pretreatment could offer protective effects for AMI through regulating the pathway of steroid hormone biosynthesis.
Collapse
Affiliation(s)
- Peng Jiang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Johansen IB, Lunde IG, Røsjø H, Christensen G, Nilsson GE, Bakken M, Overli O. Cortisol response to stress is associated with myocardial remodeling in salmonid fishes. ACTA ACUST UNITED AC 2011; 214:1313-21. [PMID: 21430209 DOI: 10.1242/jeb.053058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cardiac disease is frequently reported in farmed animals, and stress has been implicated as a factor for myocardial dysfunction in commercial fish rearing. Cortisol is a major stress hormone in teleosts, and this hormone has adverse effects on the myocardium. Strains of rainbow trout (Oncorhynchus mykiss) selected for divergent post-stress cortisol levels [high responsive (HR) and low responsive (LR)] have been established as a comparative model to examine how fish with contrasting stress-coping styles differ in their physiological and behavioral profiles. We show that the mean cardiosomatic index (CSI) of adult HR fish was 34% higher than in LR fish, mainly because of hypertrophy of the compact myocardium. To characterize the hypertrophy as physiological or pathological, we investigated specific cardiac markers at the transcriptional level. HR hearts had higher mRNA levels of cortisol receptors (MR, GR1 and GR2), increased RCAN1 levels [suggesting enhanced pro-hypertrophic nuclear factor of activated T-cell (NFAT) signaling] and increased VEGF gene expression (reflecting increased angiogenesis). Elevated collagen (Col1a2) expression and deposition in HR hearts supported enhanced fibrosis, whereas the heart failure markers ANP and BNP were not upregulated in HR hearts. To confirm our results outside the selection model, we investigated the effect of acute confinement stress in wild-type European brown trout, Salmo trutta. A positive correlation between post-stress cortisol levels and CSI was observed, supporting an association between enhanced cortisol response and myocardial remodeling. In conclusion, post-stress cortisol production correlates with myocardial remodeling, and coincides with several indicators of heart pathology, well-known from mammalian cardiology.
Collapse
Affiliation(s)
- Ida B Johansen
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences (UMB), 0476 Ås, Norway.
| | | | | | | | | | | | | |
Collapse
|
41
|
Jonker SS, Scholz TD, Segar JL. The effect of adrenalectomy on the cardiac response to subacute fetal anemia. Can J Physiol Pharmacol 2011; 89:79-88. [PMID: 21326338 DOI: 10.1139/y10-108] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The mechanisms that stimulate fetal heart growth during anemia are unknown. To examine the hypothesis that adrenal hormones contribute to this process, we determined the effects of adrenalectomy (Adx) on heart growth and the activation of cardiac mitogen-activated protein kinases (MAPKs) in the presence and absence of fetal anemia. To identify mechanisms contributing to the initiation of cardiac growth, the duration of anemia was limited to a period shorter than that previously described to result in increased cardiac mass. Four groups of fetal sheep were studied (Adx-Anemic, Adx-Control, Intact-Anemic, Intact-Control). Anemia was created by daily controlled hemorrhage for 5 days; hearts were collected for analysis at 133 d gestation (term 145 d). Cardiomyocyte morphometry, immunohistochemistry for Ki-67 (proliferation marker), and Western blotting for protein levels of MAPKs and proliferating cell nuclear antigen (PCNA) were performed. Blood pressure, heart rate, heart weight-to-body weight ratio, and cardiomyocyte length and width remained similar among groups throughout the study. PCNA levels in the Adx-Anemic group were twice as high as in any other group (both ventricles, p < 0.05). Levels of phosphorylated extracellular signal-regulated kinase (ERK) were ~60% higher in the Intact-Anemic and Adx-Anemic groups, compared with the Intact-Control and Adx-Control groups (p < 0.02). These results suggest that adrenal hormones may attenuate fetal cardiomyocyte proliferation in response to anemia (as evidenced by the increased PCNA in Adx-Anemic fetuses) and that phosphorylation of myocardial ERK results from fetal anemia, irrespective of the status of the fetal adrenal gland.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
42
|
Barel M, Perez OAB, Giozzet VA, Rafacho A, Bosqueiro JR, do Amaral SL. Exercise training prevents hyperinsulinemia, muscular glycogen loss and muscle atrophy induced by dexamethasone treatment. Eur J Appl Physiol 2009; 108:999-1007. [DOI: 10.1007/s00421-009-1272-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2009] [Indexed: 01/08/2023]
|
43
|
Beck IME, Vanden Berghe W, Vermeulen L, Yamamoto KR, Haegeman G, De Bosscher K. Crosstalk in inflammation: the interplay of glucocorticoid receptor-based mechanisms and kinases and phosphatases. Endocr Rev 2009; 30:830-82. [PMID: 19890091 PMCID: PMC2818158 DOI: 10.1210/er.2009-0013] [Citation(s) in RCA: 214] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 08/18/2009] [Indexed: 12/20/2022]
Abstract
Glucocorticoids (GCs) are steroidal ligands for the GC receptor (GR), which can function as a ligand-activated transcription factor. These steroidal ligands and derivatives thereof are the first line of treatment in a vast array of inflammatory diseases. However, due to the general surge of side effects associated with long-term use of GCs and the potential problem of GC resistance in some patients, the scientific world continues to search for a better understanding of the GC-mediated antiinflammatory mechanisms. The reversible phosphomodification of various mediators in the inflammatory process plays a key role in modulating and fine-tuning the sensitivity, longevity, and intensity of the inflammatory response. As such, the antiinflammatory GCs can modulate the activity and/or expression of various kinases and phosphatases, thus affecting the signaling efficacy toward the propagation of proinflammatory gene expression and proinflammatory gene mRNA stability. Conversely, phosphorylation of GR can affect GR ligand- and DNA-binding affinity, mobility, and cofactor recruitment, culminating in altered transactivation and transrepression capabilities of GR, and consequently leading to a modified antiinflammatory potential. Recently, new roles for kinases and phosphatases have been described in GR-based antiinflammatory mechanisms. Moreover, kinase inhibitors have become increasingly important as antiinflammatory tools, not only for research but also for therapeutic purposes. In light of these developments, we aim to illuminate the integrated interplay between GR signaling and its correlating kinases and phosphatases in the context of the clinically important combat of inflammation, giving attention to implications on GC-mediated side effects and therapy resistance.
Collapse
Affiliation(s)
- Ilse M E Beck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium
| | | | | | | | | | | |
Collapse
|
44
|
Forhead AJ, Cutts S, Matthews PA, Fowden AL. Role of thyroid hormones in the developmental control of tissue glycogen in fetal sheep near term. Exp Physiol 2009; 94:1079-87. [DOI: 10.1113/expphysiol.2009.048751] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
|
46
|
Lumbers ER, Kim MY, Burrell JH, Kumarasamy V, Boyce AC, Gibson KJ, Gatford KL, Owens JA. Effects of intrafetal IGF-I on growth of cardiac myocytes in late-gestation fetal sheep. Am J Physiol Endocrinol Metab 2009; 296:E513-9. [PMID: 19126787 DOI: 10.1152/ajpendo.90497.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intrafetal insulin-like growth factor (IGF)-I promotes cardiac hypertrophy in the late-gestation fetal sheep; whether these effects are sustained is unknown. IGF-I was infused for 4 days at 80 microg/h from 121 to 125 days of gestation, and its effects at 128 days, 3 days after the infusion stopped, were determined by comparison with untreated fetal sheep. After IGF-I treatment, fetal weights were similar to those in control fetuses but kidney weights were bigger (P < 0.05), as were spleen weights of male fetuses (P < 0.05). Cardiac myocytes were larger in female than male fetal sheep (P < 0.001). IGF-I increased male (P < 0.001) but not female myocyte volumes. IGF-I did not alter the proportions of uni- or binucleated right or left ventricular myocytes. Female fetal sheep had a greater proportion of binucleated cardiac myocytes than males (P < 0.05). IGF-I-treated fetuses had a slightly greater proportion of right ventricular nuclei in cell cycle phase G(2)/M and a reduced proportion of G(0)/G(1) phase nuclei (P < 0.1). Therefore, evidence for IGF-I-stimulated cardiac cell hyperplasia in fetal sheep in late gestation was limited. In conclusion, the greater sizes and larger proportion of binucleated cardiac myocytes in female fetal sheep suggest that myocyte maturation may occur earlier in females than in males. This may explain in part the male sex-specific responsiveness of cardiac hypertrophy to IGF-I in late gestation. If IGF-I-stimulated cardiomyocyte growth is accompanied by maturation of contractile function, IGF-I may be a potential therapeutic agent for maintaining cardiac output in preterm males.
Collapse
Affiliation(s)
- Eugenie R Lumbers
- Department of Physiology, University of New South Wales, Sydney, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Takagi S, Tanabe A, Tsuiki M, Naruse M, Takano K. Hypokalemia, diabetes mellitus, and hypercortisolemia are the major contributing factors to cardiac dysfunction in adrenal Cushing's syndrome. Endocr J 2009; 56:1009-18. [PMID: 19789419 DOI: 10.1507/endocrj.k09e-198] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Although cardiovascular complications are the major determinant of the prognosis of Cushing's syndrome (CS), factors contributing to the cardiovascular lesions are still unclear. We investigated clinical factors determining cardiac function in patients with adrenal CS. Fifty patients with adrenal CS were studied. Patients were divided into 3 groups based on their NYHA classification and electrocardiographic (ECG) findings: group A with NYHA grade 0 and normal ECG, group B with NYHA grade I and abnormal ECG, and group C with NYHA grade II or higher. Clinical and echocardiographic findings were compared between the groups. Heart failure of grade I or higher was seen in 40% and grade II or higher was seen in 8% of the patients. Age, HbA1c, and prevalence of diabetes mellitus were positively correlated and serum potassium levels were negatively correlated with the severity of cardiac dysfunction. Decreased ejection fraction (EF) and the ratio of the peak to late transmittal filling velocities (E/A), and increased left ventricular mass index (LVMI) were frequently observed. Multivariate analysis demonstrated that serum potassium and HbA1c levels were independent factors contributing to EF, while serum potassium and cortisol levels were independent factors contributing to LVMI. These results clearly demonstrated that hypokalemia, diabetes mellitus, and hypercortisolemia are the major contributing factors to cardiac dysfunction in adrenal CS. Strict control of these conditions is warranted for the prevention of cardiac dysfunction in adrenal CS.
Collapse
Affiliation(s)
- Sachiko Takagi
- Department of Medicine, Institute of Clinical Endocrinology, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | | | | | | | | |
Collapse
|
48
|
Reini SA, Wood CE, Keller-Wood M. The ontogeny of genes related to ovine fetal cardiac growth. Gene Expr Patterns 2008; 9:122-8. [PMID: 18835462 DOI: 10.1016/j.gep.2008.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Revised: 09/06/2008] [Accepted: 09/12/2008] [Indexed: 02/02/2023]
Abstract
The objective of this study was to determine the ontogenetic profiles in left and right ventricle of genes implicated in cardiac growth, including mineralocorticoid (MR) and glucocorticoid (GR) receptor, 11 beta-hydroxysteroid dehydrogenase (11beta-HSD) 1 and 2 and genes of the angiotensin system and insulin-like growth factor (IGF) family. Samples from left and right ventricles (LV, RV) were collected from hearts of sheep fetuses at 80, 100, 120, 130, and 145 days of gestation and from newborn lambs. Quantitative real-time PCR was performed to determine the MR, GR, 11beta-HSD 1 and 2, angiotensin converting enzyme (ACE) 1 and 2, IGF1, IGF2, IGF receptors IGF-1R and IGF-2R, and IGF-binding proteins (IGFBP) 2 and 3. In the LV, MR and GR both decreased toward term. In the RV, MR and GR expression did not decrease, but both 11beta-HSD 1 and 2 mRNA levels increased after birth. ACE1 expression in LV and RV sharply increases just before parturition, whereas ACE2 decreased in the LV and RV in late gestation. IGF2, IGF2R, and IGFBP2 expression levels substantially decreased in late gestation in LV and RV; IGF2R also decreased with age in LV. These patterns suggest that reduced expression of genes related to IGF and angiotensin II action occur as proliferative activity declines and terminal differentiation occurs in the late gestation fetal heart.
Collapse
Affiliation(s)
- Seth A Reini
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
49
|
Porrello ER, Widdop RE, Delbridge LMD. Early origins of cardiac hypertrophy: does cardiomyocyte attrition programme for pathological 'catch-up' growth of the heart? Clin Exp Pharmacol Physiol 2008; 35:1358-64. [PMID: 18759854 DOI: 10.1111/j.1440-1681.2008.05036.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
1. Epidemiological and experimental evidence suggests that adult development of cardiovascular disease is influenced by events of prenatal and early postnatal life. Cardiac hypertrophy is recognized as an important predictor of cardiovascular morbidity and mortality, but the developmental origins of this condition are not well understood. 2. In the heart, a switch from hyperplastic to hypertrophic cellular growth occurs during late prenatal or early postnatal life. Postnatal growth of the heart is almost entirely reliant on hypertrophy of individual cardiomyocytes, and damage to heart muscle in adulthood is typically not reparable by cell replacement. Therefore, a reduced number of cardiomyocytes may render the heart more vulnerable in situations where an increased workload is required. 3. A number of different animal models have been used to study fetal programming of adult diseases, including nutritional, hypoxic, maternal/neonatal endocrine stress and genetic models. Although studies investigating the cellular basis of myocardial disease in growth-restricted models are limited, a reduction in cardiomyocyte number through either reduced cellular proliferation or increased apoptosis appears to be a central feature. 4. The mechanisms responsible for the programming of adult cardiovascular disease are poorly understood. We hypothesize that cardiac hypertrophy can have a developmental origin in excess cardiomyocyte attrition during a critical perinatal growth window. Findings that have directly assessed the impact of fetal growth restriction on the myocardium are considered and cellular and molecular mechanisms involved in the potential pathological 'catch-up' growth of the heart during later maturation are identified.
Collapse
Affiliation(s)
- Enzo R Porrello
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
50
|
Reini SA, Dutta G, Wood CE, Keller-Wood M. Cardiac corticosteroid receptors mediate the enlargement of the ovine fetal heart induced by chronic increases in maternal cortisol. J Endocrinol 2008; 198:419-27. [PMID: 18495945 PMCID: PMC2742944 DOI: 10.1677/joe-08-0022] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Previous studies have demonstrated that modest, physiologically relevant increases in maternal cortisol in late gestation result in enlargement of the fetal heart. In this study, we investigated the role of mineralocorticoid receptor (MR) or glucocorticoid receptor (GR) in this enlargement. Ewes with single fetuses were randomly assigned at approximately 120 days of gestation to one of four groups: maternal cortisol infusion (1 mg/kg per day, cortisol); maternal cortisol infusion with fetal intrapericardial infusion of the MR antagonist (MRa) potassium canrenoate (600 microg/day; cortisol+MRa); maternal cortisol infusion with fetal intrapericardial infusion of the GR antagonist (GRa) mifepristone (50 microg/day, cortisol+GRa); and maternal saline infusion (control). At approximately 130 days of gestation, fetal heart to body weight ratio and right ventricular (RV) and left ventricular (LV) free wall thicknesses were increased in the cortisol group when compared with control group. Fetal hearts from the cortisol+MRa group weighed significantly less, with thinner LV, RV, and interventricular septum walls, when compared with the cortisol group. Fetal hearts from the cortisol+GRa group had significantly thinner RV walls than the cortisol group. Fetal arterial pressure and heart rate were not different among groups at 130 days. Picrosirius red staining of fetal hearts indicated that the increased size was not accompanied by cardiac fibrosis. These results suggest that physiologic increases in maternal cortisol in late gestation induce fetal cardiac enlargement via MR and, to a lesser extent, by GR, and indicate that the enlargement is not secondary to an increase in fetal blood pressure or an increase in fibrosis within the fetal heart.
Collapse
Affiliation(s)
- Seth A. Reini
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL
| | - Garima Dutta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL
| | - Charles E. Wood
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL
| |
Collapse
|