1
|
Lock MC, Patey OV, Smith KLM, Niu Y, Jaggs B, Trafford AW, Giussani DA, Galli GLJ. Maladaptive cardiomyocyte calcium handling in adult offspring of hypoxic pregnancy: protection by antenatal maternal melatonin. J Physiol 2024; 602:6683-6703. [PMID: 39572933 DOI: 10.1113/jp287325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/10/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic fetal hypoxia is one of the most common complications of pregnancy and can programme cardiac abnormalities in adult offspring including ventricular remodelling, diastolic dysfunction and sympathetic dominance. However, the underlying mechanisms at the level of the cardiomyocyte are unknown, preventing the identification of targets for therapeutic intervention. Therefore, we aimed to link echocardiographic data with cardiomyocyte function to reveal cellular mechanism for cardiac dysfunction in rat offspring from hypoxic pregnancy. Further, we investigated the potential of maternal treatment with melatonin as antenatal antioxidant therapy. Wistar rats were randomly allocated into normoxic (21% O2) or hypoxic (13% O2) pregnancy with or without melatonin treatment (5 µg/ml; normoxic melatonin in the maternal drinking water from gestational day 6 to 20 (term = 22 days). After delivery, male and female offspring were maintained to adulthood (16 weeks). Cardiomyocytes were isolated from the left and right ventricles, and calcium (Ca2+) handling was investigated in field-stimulated myocytes. Systolic and diastolic function was negatively impacted in male and female offspring of hypoxic pregnancy demonstrating biventricular systolic and diastolic dysfunction and compensatory increases in cardiac output. Ca2+ transients from isolated cardiomyocytes in offspring of both sexes in hypoxic pregnancy displayed diastolic dysfunction with a reduced rate of [Ca2+]i recovery. Cardiac and cardiomyocyte dysfunction in male and female adult offspring was ameliorated by maternal antenatal treatment with melatonin in hypoxic pregnancy. Therefore, cardiomyocyte Ca2+ mishandling provides a cellular mechanism explaining functional deficits in hearts of male and female offspring in pregnancies complicated by chronic fetal hypoxia. KEY POINTS: This study identified significant changes in Ca2+ handling within cardiomyocytes isolated from offspring of hypoxic pregnancy including reduced systolic Ca2+ transients, impaired diastolic recovery of [Ca2+]i and a greater increase in systolic [Ca2+]i amplitude to β-adrenergic stimulation. These changes in cardiomyocyte Ca2+ handling help to explain dysregulation of biventricular systolic and diastolic dysfunction determined by echocardiography. The data show protection against maladaptive cardiomyocyte calcium handling and thereby improvement in cardiac function in adult offspring of hypoxic pregnancy treated with melatonin with doses lower than those recommended for overcoming jet lag in humans. Melatonin treatment alone in healthy pregnancy did cause some alterations in cardiac structure. Therefore, maternal treatment with melatonin should only be given to pregnancies affected by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Mitchell C Lock
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Olga V Patey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kerri L M Smith
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ben Jaggs
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gina L J Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Warrington JP, Collins HE, Davidge ST, do Carmo JM, Goulopoulou S, Intapad S, Loria AS, Sones JL, Wold LE, Zinkhan EK, Alexander BT. Guidelines for in vivo models of developmental programming of cardiovascular disease risk. Am J Physiol Heart Circ Physiol 2024; 327:H221-H241. [PMID: 38819382 PMCID: PMC11380980 DOI: 10.1152/ajpheart.00060.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Research using animals depends on the generation of offspring for use in experiments or for the maintenance of animal colonies. Although not considered by all, several different factors preceding and during pregnancy, as well as during lactation, can program various characteristics in the offspring. Here, we present the most common models of developmental programming of cardiovascular outcomes, important considerations for study design, and provide guidelines for producing and reporting rigorous and reproducible cardiovascular studies in offspring exposed to normal conditions or developmental insult. These guidelines provide considerations for the selection of the appropriate animal model and factors that should be reported to increase rigor and reproducibility while ensuring transparent reporting of methods and results.
Collapse
Grants
- 20YVNR35490079 American Heart Association (AHA)
- R01HL139348 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135158 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54GM115428 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01AG057046 HHS | NIH | National Institute on Aging (NIA)
- P20 GM104357 NIGMS NIH HHS
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 GM149404 NIGMS NIH HHS
- P20GM104357 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM135002 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL163003 NHLBI NIH HHS
- R01HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01DK121411 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Excellence Faculty Support Grant Jewish Heritage Fund
- P30GM149404 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P30GM14940 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- 23SFRNPCS1067044 American Heart Association (AHA)
- R01 HL146562 NHLBI NIH HHS
- R56HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54 GM115428 NIGMS NIH HHS
- 1R01HL163076 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL51971 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- FS154313 CIHR
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Helen E Collins
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, Kentucky, United States
| | - Sandra T Davidge
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jussara M do Carmo
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, United States
- Department of Gynecology, and Obstetrics, Loma Linda University, Loma Linda, California, United States
| | - Suttira Intapad
- Department of Pharmacology, Tulane University, New Orleans, Louisiana, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jenny L Sones
- Equine Reproduction Laboratory, Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Erin K Zinkhan
- Department of Pediatrics, University of Utah and Intermountain Health, Salt Lake City, Utah, United States
- Intermountain Health, Salt Lake City, Utah, United States
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
3
|
Collins HE, Alexander BT, Care AS, Davenport MH, Davidge ST, Eghbali M, Giussani DA, Hoes MF, Julian CG, LaVoie HA, Olfert IM, Ozanne SE, Bytautiene Prewit E, Warrington JP, Zhang L, Goulopoulou S. Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum. Am J Physiol Heart Circ Physiol 2024; 327:H191-H220. [PMID: 38758127 PMCID: PMC11380979 DOI: 10.1152/ajpheart.00055.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.
Collapse
Grants
- HL169157 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HD088590 NICHD NIH HHS
- HD083132 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- The Biotechnology and Biological Sciences Research Council
- P20GM103499 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- British Heart Foundation (BHF)
- R21 HD111908 NICHD NIH HHS
- Distinguished University Professor
- The Lister Insititute
- ES032920 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL149608 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Royal Society (The Royal Society)
- U.S. Department of Defense (DOD)
- HL138181 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- MC_00014/4 UKRI | Medical Research Council (MRC)
- RG/17/8/32924 British Heart Foundation
- Jewish Heritage Fund for Excellence
- HD111908 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP2002129 NHMRC Ideas Grant
- HL159865 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL131182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NS103017 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL138181 NHLBI NIH HHS
- 20CSA35320107 American Heart Association (AHA)
- RG/17/12/33167 British Heart Foundation (BHF)
- National Heart Foundation Future Leader Fellowship
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155295 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HD088590-06 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- WVU SOM Synergy Grant
- R01 HL146562 NHLBI NIH HHS
- R01 HL159865 NHLBI NIH HHS
- Canadian Insitute's of Health Research Foundation Grant
- R01 HL169157 NHLBI NIH HHS
- HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ES034646-01 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL150472 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 2021T017 Dutch Heart Foundation Dekker Grant
- MC_UU_00014/4 Medical Research Council
- R01 HL163003 NHLBI NIH HHS
- Christenson professor In Active Healthy Living
- National Heart Foundation
- Dutch Heart Foundation Dekker
- WVU SOM Synergy
- Jewish Heritage
- Department of Health | National Health and Medical Research Council (NHMRC)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Helen E Collins
- University of Louisville, Louisville, Kentucky, United States
| | - Barbara T Alexander
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alison S Care
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | - Mansoureh Eghbali
- University of California Los Angeles, Los Angeles, California, United States
| | | | | | - Colleen G Julian
- University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Holly A LaVoie
- University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - I Mark Olfert
- West Virginia University School of Medicine, Morgantown, West Virginia, United States
| | | | | | - Junie P Warrington
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Lubo Zhang
- Loma Linda University School of Medicine, Loma Linda, California, United States
| | | |
Collapse
|
4
|
Kamenshchyk A, Belenichev I, Oksenych V, Kamyshnyi O. Combined Pharmacological Modulation of Translational and Transcriptional Activity Signaling Pathways as a Promising Therapeutic Approach in Children with Myocardial Changes. Biomolecules 2024; 14:477. [PMID: 38672493 PMCID: PMC11047929 DOI: 10.3390/biom14040477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial hypertrophy is the most common condition that accompanies heart development in children. Transcriptional gene expression regulating pathways play a critical role both in cardiac embryogenesis and in the pathogenesis of congenital hypertrophic cardiomyopathy, neonatal posthypoxic myocardial hypertrophy, and congenital heart diseases. This paper describes the state of cardiac gene expression and potential pharmacological modulators at different transcriptional levels. An experimental model of perinatal cardiac hypoxia showed the downregulated expression of genes responsible for cardiac muscle integrity and overexpressed genes associated with energy metabolism and apoptosis, which may provide a basis for a therapeutic approach. Current evidence suggests that RNA drugs, theaflavin, neuraminidase, proton pumps, and histone deacetylase inhibitors are promising pharmacological agents in progressive cardiac hypertrophy. The different points of application of the above drugs make combined use possible, potentiating the effects of inhibition in specific signaling pathways. The special role of N-acetyl cysteine in both the inhibition of several signaling pathways and the reduction of oxidative stress was emphasized.
Collapse
Affiliation(s)
- Andrii Kamenshchyk
- Department of Hospital Pediatrics, Zaporizhzhya State Medical and Pharmaceutical University, 69035 Zaporizhzhya, Ukraine
| | - Igor Belenichev
- Department of Pharmacology, Zaporizhzhya State Medical and Pharmaceutical University, 69035 Zaporizhzhya, Ukraine;
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
5
|
Wang Z, Camm EJ, Nuzzo AM, Spiroski AM, Skeffington KL, Ashmore TJ, Rolfo A, Todros T, Logan A, Ma J, Murphy MP, Niu Y, Giussani DA. In vivo mitochondria-targeted protection against uterine artery vascular dysfunction and remodelling in rodent hypoxic pregnancy. J Physiol 2024; 602:1211-1225. [PMID: 38381050 DOI: 10.1113/jp286178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
Gestational hypoxia adversely affects uterine artery function, increasing complications. However, an effective therapy remains unidentified. Here, we show in rodent uterine arteries that hypoxic pregnancy promotes hypertrophic remodelling, increases constrictor reactivity via protein kinase C signalling, and triggers compensatory dilatation via nitric oxide-dependent mechanisms and stimulation of large conductance Ca2+ -activated K+ -channels. Maternal in vivo oral treatment with the mitochondria-targeted antioxidant MitoQ in hypoxic pregnancy normalises uterine artery reactivity and prevents vascular remodelling. From days 6-20 of gestation (term ∼22 days), female Wistar rats were randomly assigned to normoxic or hypoxic (13-14% O2 ) pregnancy ± daily maternal MitoQ treatment (500 µm in drinking water). At 20 days of gestation, maternal, placental and fetal tissue was frozen to determine MitoQ uptake. The uterine arteries were harvested and, in one segment, constrictor and dilator reactivity was determined by wire myography. Another segment was fixed for unbiased stereological analysis of vessel morphology. Maternal administration of MitoQ in both normoxic and hypoxic pregnancy crossed the placenta and was present in all tissues analysed. Hypoxia increased uterine artery constrictor responses to norepinephrine, angiotensin II and the protein kinase C activator, phorbol 12,13-dibutyrate. Hypoxia enhanced dilator reactivity to sodium nitroprusside, the large conductance Ca2+ -activated K+ -channel activator NS1619 and ACh via increased nitric oxide-dependent mechanisms. Uterine arteries from hypoxic pregnancy showed increased wall thickness and MitoQ treatment in hypoxic pregnancy prevented all effects on uterine artery reactivity and remodelling. The data support mitochondria-targeted therapy against adverse changes in uterine artery structure and function in high-risk pregnancy. KEY POINTS: Dysfunction and remodelling of the uterine artery are strongly implicated in many pregnancy complications, including advanced maternal age, maternal hypertension of pregnancy, maternal obesity, gestational diabetes and pregnancy at high altitude. Such complications not only have immediate adverse effects on the growth of the fetus, but also they can also increase the risk of cardiovascular disease in the mother and offspring. Despite this, there is a significant unmet clinical need for therapeutics that treat uterine artery vascular dysfunction in adverse pregnancy. Here, we show in a rodent model of gestational hypoxia that in vivo oral treatment of the mitochondria-targeted antioxidant MitoQ protects against uterine artery vascular dysfunction and remodelling, supporting the use of mitochondria-targeted therapy against adverse changes in uterine artery structure and function in high-risk pregnancy.
Collapse
Affiliation(s)
- Zhongchao Wang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Anna Maria Nuzzo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Katie L Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Tullia Todros
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Angela Logan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| |
Collapse
|
6
|
Popazova O, Belenichev I, Bukhtiyarova N, Ryzhenko V, Oksenych V, Kamyshnyi A. Cardioprotective Activity of Pharmacological Agents Affecting NO Production and Bioavailability in the Early Postnatal Period after Intrauterine Hypoxia in Rats. Biomedicines 2023; 11:2854. [PMID: 37893227 PMCID: PMC10604160 DOI: 10.3390/biomedicines11102854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Intrauterine hypoxia in newborns leads to a multifaceted array of alterations that exert a detrimental impact on the cardiovascular system. The aim of this research was to assess the cardioprotective effects of modulators of the nitric oxide (NO) system, including L-arginine, Thiotriazoline, Angiolin, and Mildronate, during the early postnatal period following intrauterine hypoxia. Methods: The study involved 50 female white rats. Pregnant female rats were given a daily intraperitoneal dose of 50 mg/kg of sodium nitrite starting on the 16th day of pregnancy. A control group of pregnant rats received saline instead. The resulting offspring were divided into the following groups: Group 1-intact rats; Group 2-rat pups subjected to prenatal hypoxia (PH) and daily treated with physiological saline; and Groups 3 to 6-rat pups exposed to prenatal hypoxia and treated daily from the 1st to the 30th day after birth. Nitrotyrosine levels, eNOS, iNOS, and NO metabolites were evaluated using ELISA; to measure the expression levels of iNOS mRNA and eNOS mRNA, a PCR test was utilized. Results: Angiolin enhances the expression of eNOS mRNA and boosts eNOS activity in the myocardium of rats with ischemic conditions. Arginine and particularly Thiotriazoline exhibited a consistent impact in restoring normal parameters of the cardiac nitroxidergic system following PH. Mildronate notably raised iNOS mRNA levels and notably reduced nitrotyrosine levels, providing further support for its antioxidative characteristics.
Collapse
Affiliation(s)
- Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Victor Ryzhenko
- Department of Medical and Pharmaceutical Informatics and Advanced Technologies, Zaporizhzhia State Medical University, 69000 Zaporizhzhia, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine
| |
Collapse
|
7
|
Chatterjee P, Holody CD, Kirschenman R, Graton ME, Spaans F, Phillips TJ, Case CP, Bourque SL, Lemieux H, Davidge ST. Sex-Specific Effects of Prenatal Hypoxia and a Placental Antioxidant Treatment on Cardiac Mitochondrial Function in the Young Adult Offspring. Int J Mol Sci 2023; 24:13624. [PMID: 37686430 PMCID: PMC10487956 DOI: 10.3390/ijms241713624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Prenatal hypoxia is associated with placental oxidative stress, leading to impaired fetal growth and an increased risk of cardiovascular disease in the adult offspring; however, the mechanisms are unknown. Alterations in mitochondrial function may result in impaired cardiac function in offspring. In this study, we hypothesized that cardiac mitochondrial function is impaired in adult offspring exposed to intrauterine hypoxia, which can be prevented by placental treatment with a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ). Cardiac mitochondrial respiration was assessed in 4-month-old rat offspring exposed to prenatal hypoxia (11% O2) from gestational day (GD)15-21 receiving either saline or nMitoQ on GD 15. Prenatal hypoxia did not alter cardiac mitochondrial oxidative phosphorylation capacity in the male offspring. In females, the NADH + succinate pathway capacity decreased by prenatal hypoxia and tended to be increased by nMitoQ. Prenatal hypoxia also decreased the succinate pathway capacity in females. nMitoQ treatment increased respiratory coupling efficiency in prenatal hypoxia-exposed female offspring. In conclusion, prenatal hypoxia impaired cardiac mitochondrial function in adult female offspring only, which was improved with prenatal nMitoQ treatment. Therefore, treatment strategies targeting placental oxidative stress in prenatal hypoxia may reduce the risk of cardiovascular disease in adult offspring by improving cardiac mitochondrial function in a sex-specific manner.
Collapse
Affiliation(s)
- Paulami Chatterjee
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Claudia D. Holody
- Faculty Saint-Jean, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Murilo E. Graton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Tom J. Phillips
- UK Dementia Research Institute, Cardiff University, Cardiff CF10 3AT, UK;
| | - C. Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol BS10 5NB, UK;
| | - Stephane L. Bourque
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Hélène Lemieux
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
- Faculty Saint-Jean, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sandra T. Davidge
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| |
Collapse
|
8
|
Lock MC, Botting KJ, Allison BJ, Niu Y, Ford SG, Murphy MP, Orgeig S, Giussani DA, Morrison JL. MitoQ as an antenatal antioxidant treatment improves markers of lung maturation in healthy and hypoxic pregnancy. J Physiol 2023; 601:3647-3665. [PMID: 37467062 PMCID: PMC10952154 DOI: 10.1113/jp284786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023] Open
Abstract
Chronic fetal hypoxaemia is a common pregnancy complication that increases the risk of infants experiencing respiratory complications at birth. In turn, chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in animal models of hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. However, whether antenatal antioxidant therapy confers any benefit on lung development in complicated pregnancies has not yet been investigated. Here, we tested the hypothesis that maternal antenatal treatment with MitoQ will protect the developing lung in hypoxic pregnancy in sheep, a species with similar fetal lung developmental milestones as humans. Maternal treatment with MitoQ during late gestation promoted fetal pulmonary surfactant maturation and an increase in the expression of lung mitochondrial complexes III and V independent of oxygenation. Maternal treatment with MitoQ in hypoxic pregnancy also increased the expression of genes regulating liquid reabsorption in the fetal lung. These data support the hypothesis tested and suggest that MitoQ as an antenatal targeted antioxidant treatment may improve lung maturation in the late gestation fetus. KEY POINTS: Chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. MitoQ is a targeted antioxidant that uses the cell and the mitochondrial membrane potential to accumulate within the mitochondria. Treatment of healthy or hypoxic pregnancy with MitoQ, increases the expression of key molecules involved in surfactant maturation, lung liquid reabsorption and in mitochondrial proteins driving ATP synthesis in the fetal sheep lung. There were no detrimental effects of MitoQ treatment alone on the molecular components measured in the present study, suggesting that maternal antioxidant treatment has no effect on other components of normal maturation of the surfactant system.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Beth J. Allison
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Sage G. Ford
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sandra Orgeig
- UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Dino A. Giussani
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
9
|
Garrud TAC, Teulings NEWD, Niu Y, Skeffington KL, Beck C, Itani N, Conlon FG, Botting KJ, Nicholas LM, Tong W, Derks JB, Ozanne SE, Giussani DA. Molecular mechanisms underlying adverse effects of dexamethasone and betamethasone in the developing cardiovascular system. FASEB J 2023; 37:e22887. [PMID: 37132324 PMCID: PMC10946807 DOI: 10.1096/fj.202200676rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 05/04/2023]
Abstract
Antenatal glucocorticoids accelerate fetal lung maturation and reduce mortality in preterm babies but can trigger adverse effects on the cardiovascular system. The mechanisms underlying off-target effects of the synthetic glucocorticoids mostly used, Dexamethasone (Dex) and Betamethasone (Beta), are unknown. We investigated effects of Dex and Beta on cardiovascular structure and function, and underlying molecular mechanism using the chicken embryo, an established model system to isolate effects of therapy on the developing heart and vasculature, independent of effects on the mother or placenta. Fertilized eggs were treated with Dex (0.1 mg kg-1 ), Beta (0.1 mg kg-1 ), or water vehicle (Control) on embryonic day 14 (E14, term = 21 days). At E19, biometry, cardiovascular function, stereological, and molecular analyses were determined. Both glucocorticoids promoted growth restriction, with Beta being more severe. Beta compared with Dex induced greater cardiac diastolic dysfunction and also impaired systolic function. While Dex triggered cardiomyocyte hypertrophy, Beta promoted a decrease in cardiomyocyte number. Molecular changes of Dex on the developing heart included oxidative stress, activation of p38, and cleaved caspase 3. In contrast, impaired GR downregulation, activation of p53, p16, and MKK3 coupled with CDK2 transcriptional repression linked the effects of Beta on cardiomyocyte senescence. Beta but not Dex impaired NO-dependent relaxation of peripheral resistance arteries. Beta diminished contractile responses to potassium and phenylephrine, but Dex enhanced peripheral constrictor reactivity to endothelin-1. We conclude that Dex and Beta have direct differential detrimental effects on the developing cardiovascular system.
Collapse
Affiliation(s)
- Tessa A. C. Garrud
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Noor E. W. D. Teulings
- Institute of Metabolic Science‐Metabolic Research Laboratories, MRC Metabolic Diseases UnitUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Katie L. Skeffington
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Christian Beck
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Nozomi Itani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Fiona G. Conlon
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Kimberley J. Botting
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Lisa M. Nicholas
- Institute of Metabolic Science‐Metabolic Research Laboratories, MRC Metabolic Diseases UnitUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
| | - Wen Tong
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jan B. Derks
- Department of Perinatal MedicineUniversity Medical CentreUtrechtNetherlands
| | - Susan E. Ozanne
- Institute of Metabolic Science‐Metabolic Research Laboratories, MRC Metabolic Diseases UnitUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
- BHF Cardiovascular Centre for Research ExcellenceUniversity of CambridgeCambridgeUK
- Strategic Research Initiative in ReproductionUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Dino A. Giussani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- BHF Cardiovascular Centre for Research ExcellenceUniversity of CambridgeCambridgeUK
- Strategic Research Initiative in ReproductionUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| |
Collapse
|
10
|
Kane AD, Herrera EA, Niu Y, Camm EJ, Allison BJ, Tijsseling D, Lusby C, Derks JB, Brain KL, Bronckers IM, Cross CM, Berends L, Giussani DA. Combined Statin and Glucocorticoid Therapy for the Safer Treatment of Preterm Birth. Hypertension 2023; 80:837-851. [PMID: 36724801 PMCID: PMC10017302 DOI: 10.1161/hypertensionaha.122.19647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 01/03/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND Prematurity is strongly associated with poor respiratory function in the neonate. Rescue therapies include treatment with glucocorticoids due to their anti-inflammatory and maturational effects on the developing lung. However, glucocorticoid treatment in the infant can increase the risk of long-term cardiovascular complications including hypertension, cardiac, and endothelial dysfunction. Accumulating evidence implicates a molecular link between glucocorticoid excess and depletion of nitric oxide (NO) bioavailability as a mechanism underlying the detrimental effects of postnatal steroids on the heart and circulation. Therefore, combined glucocorticoid and statin therapy, by increasing NO bioavailability, may protect the developing cardiovascular system while maintaining beneficial effects on the lung. METHODS We investigated combined glucocorticoid and statin therapy using an established rodent model of prematurity and combined experiments of cardiovascular function in vivo, with those in isolated organs as well as measurements at the cellular and molecular levels. RESULTS We show that neonatal glucocorticoid treatment increases the risk of later cardiovascular dysfunction in the offspring. Underlying mechanisms include decreased circulating NO bioavailability, sympathetic hyper-reactivity, and NO-dependent endothelial dysfunction. Combined neonatal glucocorticoid and statin therapy protects the developing cardiovascular system by normalizing NO and sympathetic signaling, without affecting pulmonary maturational or anti-inflammatory effects of glucocorticoids. CONCLUSIONS Therefore, combined glucocorticoid and statin therapy may be safer than glucocorticoids alone for the treatment of preterm birth.
Collapse
Affiliation(s)
- Andrew D. Kane
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
| | - Emilio A. Herrera
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile (E.A.H.)
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
- The Cambridge BHF Centre for Research Excellence, Cambridge, United Kingdom (Y.N., D.A.G.)
- The Cambridge Strategic Research Initiative in Reproduction, Cambridge, United Kingdom (Y.N., D.A.G.)
| | - Emily J. Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (E.J.C., B.J.A.)
| | - Beth J. Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (E.J.C., B.J.A.)
| | - Deodata Tijsseling
- Perinatal Center, University Medical Center, Utrecht, the Netherlands (D.T., J.B.D.)
| | - Ciara Lusby
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
| | - Jan B. Derks
- Perinatal Center, University Medical Center, Utrecht, the Netherlands (D.T., J.B.D.)
| | - Kirsty L. Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
| | - Inge M. Bronckers
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, the Netherlands (I.M.B.)
| | - Christine M. Cross
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
| | - Lindsey Berends
- Institute of Metabolic Science, University of Cambridge Metabolic Research Laboratories, Addenbrooke’s Hospital, Cambridge, United Kingdom (L.B.)
| | - Dino A. Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (A.D.K., E.A.H., Y.N., E.J.C., B.J.A., C.L., K.L.B., C.M.C., D.A.G.)
- The Cambridge BHF Centre for Research Excellence, Cambridge, United Kingdom (Y.N., D.A.G.)
- The Cambridge Strategic Research Initiative in Reproduction, Cambridge, United Kingdom (Y.N., D.A.G.)
| |
Collapse
|
11
|
The Long-Term Effects of Prenatal Hypoxia on Coronary Artery Function of the Male and Female Offspring. Biomedicines 2022; 10:biomedicines10123019. [PMID: 36551775 PMCID: PMC9776081 DOI: 10.3390/biomedicines10123019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Prenatal hypoxia predisposes the offspring to the development of cardiovascular (CV) dysfunction in adult life. Using a rat model, we assessed the effect of prenatal hypoxia on vasoconstrictive and vasodilative mechanisms in left anterior descending coronary arteries of 4- and 9.5-month-old offspring. Endothelium-dependent relaxation to methylcholine and vasoconstriction responses to endothelin-1 (ET-1) were assessed by wire myography. Prenatal hypoxia impaired endothelium-dependent vasodilation in 4- and 9.5-month-old offspring. Inhibition of nitric oxide (NO) synthase prevented coronary artery relaxation in all groups. Inhibition of prostaglandin H synthase (PGHS) improved relaxation in prenatally hypoxic males and tended to improve vasorelaxation in females, suggesting that impaired vasodilation was mediated via increased PGHS-dependent vasoconstriction. An enhanced contribution of endothelium-dependent hyperpolarization to coronary artery vasodilation was observed in prenatally hypoxic males and females. No changes in endothelial NO synthase (eNOS) and PGHS-1 expressions were observed, while PGHS-2 expression was decreased in only prenatally hypoxic males. At 4 months, ET-1 responses were similar between groups, while ETB inhibition (with BQ788) tended to decrease ET-1-mediated responses in only prenatally hypoxic females. At 9.5 months, ET-1-mediated responses were decreased in only prenatally hypoxic females. Our data suggest that prenatal hypoxia has long-term similar effects on the mechanisms of impaired endothelium-dependent vasodilation in coronary arteries from adult male and female offspring; however, coronary artery contractile capacity is impaired only in prenatally hypoxic females. Understanding the mechanistic pathways involved in the programming of CV disease may allow for the development of therapeutic interventions.
Collapse
|
12
|
Rodríguez-Rovira I, Arce C, De Rycke K, Pérez B, Carretero A, Arbonés M, Teixidò-Turà G, Gómez-Cabrera MC, Campuzano V, Jiménez-Altayó F, Egea G. Allopurinol blocks aortic aneurysm in a mouse model of Marfan syndrome via reducing aortic oxidative stress. Free Radic Biol Med 2022; 193:538-550. [PMID: 36347404 DOI: 10.1016/j.freeradbiomed.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Increasing evidence indicates that redox stress participates in MFS aortopathy, though its mechanistic contribution is little known. We reported elevated reactive oxygen species (ROS) formation and NADPH oxidase NOX4 upregulation in MFS patients and mouse aortae. Here we address the contribution of xanthine oxidoreductase (XOR), which catabolizes purines into uric acid and ROS in MFS aortopathy. METHODS AND RESULTS In aortic samples from MFS patients, XOR protein expression, revealed by immunohistochemistry, increased in both the tunicae intima and media of the dilated zone. In MFS mice (Fbn1C1041G/+), aortic XOR mRNA transcripts and enzymatic activity of the oxidase form (XO) were augmented in the aorta of 3-month-old mice but not in older animals. The administration of the XOR inhibitor allopurinol (ALO) halted the progression of aortic root aneurysm in MFS mice. ALO administrated before the onset of the aneurysm prevented its subsequent development. ALO also inhibited MFS-associated endothelial dysfunction as well as elastic fiber fragmentation, nuclear translocation of pNRF2 and increased 3'-nitrotyrosine levels, and collagen maturation remodeling, all occurring in the tunica media. ALO reduced the MFS-associated large aortic production of H2O2, and NOX4 and MMP2 transcriptional overexpression. CONCLUSIONS Allopurinol interferes in aortic aneurysm progression acting as a potent antioxidant. This study strengthens the concept that redox stress is an important determinant of aortic aneurysm formation and progression in MFS and warrants the evaluation of ALO therapy in MFS patients.
Collapse
Affiliation(s)
- Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Cristina Arce
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Karo De Rycke
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Belén Pérez
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Aitor Carretero
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Marc Arbonés
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Gisela Teixidò-Turà
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER-CV, Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - Mari Carmen Gómez-Cabrera
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain.
| |
Collapse
|
13
|
Smith KLM, Swiderska A, Lock MC, Graham L, Iswari W, Choudhary T, Thomas D, Kowash HM, Desforges M, Cottrell EC, Trafford AW, Giussani DA, Galli GLJ. Chronic developmental hypoxia alters mitochondrial oxidative capacity and reactive oxygen species production in the fetal rat heart in a sex-dependent manner. J Pineal Res 2022; 73:e12821. [PMID: 35941749 PMCID: PMC9540814 DOI: 10.1111/jpi.12821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Abstract
Insufficient oxygen supply (hypoxia) during fetal development leads to cardiac remodeling and a predisposition to cardiovascular disease in later life. Previous work has shown hypoxia causes oxidative stress in the fetal heart and alters the activity and expression of mitochondrial proteins in a sex-dependent manner. However, the functional effects of these modifications on mitochondrial respiration remain unknown. Furthermore, while maternal antioxidant treatments are emerging as a promising new strategy to protect the hypoxic fetus, whether these treatments convey similar protection to cardiac mitochondria in the male or female fetus has not been investigated. Therefore, using an established rat model, we measured the sex-dependent effects of gestational hypoxia and maternal melatonin treatment on fetal cardiac mitochondrial respiration, reactive oxygen species (ROS) production, and lipid peroxidation. Pregnant Wistar rats were subjected to normoxia or hypoxia (13% oxygen) during gestational days (GDs) 6-20 (term ~22 days) with or without melatonin treatment (5 µg/ml in maternal drinking water). On GD 20, mitochondrial aerobic respiration and H2 O2 production were measured in fetal heart tissue, together with lipid peroxidation and citrate synthase (CS) activity. Gestational hypoxia reduced maternal body weight gain (p < .01) and increased placental weight (p < .05) but had no effect on fetal weight or litter size. Cardiac mitochondria from male but not female fetuses of hypoxic pregnancy had reduced respiratory capacity at Complex II (CII) (p < .05), and an increase in H2 O2 production/O2 consumption (p < .05) without any changes in lipid peroxidation. CS activity was also unchanged in both sexes. Despite maternal melatonin treatment increasing maternal and fetal plasma melatonin concentration (p < .001), melatonin treatment had no effect on any of the mitochondrial parameters investigated. To conclude, we show that gestational hypoxia leads to ROS generation from the mitochondrial electron transport chain and affects fetal cardiac mitochondrial respiration in a sex-dependent manner. We also show that maternal melatonin treatment had no effect on these relationships, which has implications for the development of future therapies for hypoxic pregnancies.
Collapse
Affiliation(s)
- Kerri L. M. Smith
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Agnieszka Swiderska
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Mitchell C. Lock
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Lucia Graham
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Wulan Iswari
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Tashi Choudhary
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Donna Thomas
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Hager M. Kowash
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Michelle Desforges
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Elizabeth C. Cottrell
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Andrew W. Trafford
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Dino A. Giussani
- Department of Physiology Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Gina L. J. Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
14
|
Hula N, Vu J, Quon A, Kirschenman R, Spaans F, Liu R, Cooke CLM, Davidge ST. Sex-Specific Effects of Prenatal Hypoxia on the Cardiac Endothelin System in Adult Offspring. Am J Physiol Heart Circ Physiol 2022; 322:H442-H450. [PMID: 35119336 DOI: 10.1152/ajpheart.00636.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal hypoxia, a major consequence of complicated pregnancies, impairs offspring cardiac tolerance to ischemia/reperfusion (I/R) insult, however, the mechanisms remain unknown. Endothelin-1 (ET-1) signaling through the endothelin A receptors (ETA) is associated with cardiac dysfunction. We hypothesized that prenatal hypoxia exacerbates cardiac susceptibility to I/R via increased ET-1 and ETA levels, while ETA inhibition ameliorates this. Pregnant Sprague-Dawley rats were exposed to normoxia (21% O2) or hypoxia (11% O2) on gestational days 15-21. Offspring were aged to 4 months, and hearts were aerobically perfused or subjected to ex vivo I/R, with or without pre-infusion with an ETA antagonist (ABT-627). ET-1 levels were assessed with ELISA in aerobically perfused and post-I/R left ventricles (LV). ETA and ETB levels were assessed by Western blotting in non-perfused LV. As hypothesized, ABT-627 infusion tended to improve post-I/R recovery in hypoxic females (p=0.0528), however, surprisingly, ABT-627 prevented post-I/R recovery only in the hypoxic males (p<0.001). ET-1 levels were increased in post-I/R LV in both sexes regardless of the prenatal exposure (p<0.01). ETA expression was similar among all groups, while ETB (isoform C) levels were decreased in prenatally hypoxic females (p<0.05). In prenatally hypoxic males, ETA signaling may be essential for tolerance to I/R, while in prenatally hypoxic females, ETA may contribute to cardiac dysfunction. Our data illustrate that understanding the prenatal history has critical implications for treatment strategies in adult chronic diseases.
Collapse
Affiliation(s)
- Nataliia Hula
- Department of Physiology, University of Alberta, Edmonton, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jennie Vu
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Ricky Liu
- Department of Physiology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Aguilera N, Salas-Pérez F, Ortíz M, Álvarez D, Echiburú B, Maliqueo M. Rodent models in placental research. Implications for fetal origins of adult disease. Anim Reprod 2022; 19:e20210134. [PMID: 35493783 PMCID: PMC9037606 DOI: 10.1590/1984-3143-ar2021-0134] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/21/2022] [Indexed: 11/22/2022] Open
|
16
|
Lakshman R, Spiroski AM, McIver LB, Murphy MP, Giussani DA. Noninvasive Biomarkers for Cardiovascular Dysfunction Programmed in Male Offspring of Adverse Pregnancy. Hypertension 2021; 78:1818-1828. [PMID: 34757774 PMCID: PMC8577293 DOI: 10.1161/hypertensionaha.121.17926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rama Lakshman
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom
| | - Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom
| | - Lauren B McIver
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom
| | - Michael P Murphy
- MRC Mitochondria Biology Unit (M.P.M.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Department of Medicine (M.P.M., D.A.G.), University of Cambridge, United Kingdom
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Department of Medicine (M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge Strategic Research Initiative in Reproduction, United Kingdom (D.A.G.)
| |
Collapse
|
17
|
Abstract
Heart disease remains one of the greatest killers. In addition to genetics and traditional lifestyle risk factors, we now understand that adverse conditions during pregnancy can also increase susceptibility to cardiovascular disease in the offspring. Therefore, the mechanisms by which this occurs and possible preventative therapies are of significant contemporary interest to the cardiovascular community. A common suboptimal pregnancy condition is a sustained reduction in fetal oxygenation. Chronic fetal hypoxia results from any pregnancy with increased placental vascular resistance, such as in preeclampsia, placental infection, or maternal obesity. Chronic fetal hypoxia may also arise during pregnancy at high altitude or because of maternal respiratory disease. This article reviews the short- and long-term effects of hypoxia on the fetal cardiovascular system, and the importance of chronic fetal hypoxia in triggering a developmental origin of future heart disease in the adult progeny. The work summarizes evidence derived from human studies as well as from rodent, avian, and ovine models. There is a focus on the discovery of the molecular link between prenatal hypoxia, oxidative stress, and increased cardiovascular risk in adult offspring. Discussion of mitochondria-targeted antioxidant therapy offers potential targets for clinical intervention in human pregnancy complicated by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Dino A Giussani
- Department of Physiology, Development, and Neuroscience; The Barcroft Centre; Cambridge Cardiovascular British Heart Foundation Centre for Research Excellence; and Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, UK
| |
Collapse
|
18
|
Li S, Chen Y, Zhang Y, Qiu F, Zeng F, Shi L. Prenatal exercise reprograms the development of hypertension progress and improves vascular health in SHR offspring. Vascul Pharmacol 2021; 139:106885. [PMID: 34116258 DOI: 10.1016/j.vph.2021.106885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 05/12/2021] [Accepted: 06/06/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Upregulation of L-type voltage-gated Ca2+ (CaV1.2) channel in the arterial myocytes is a hallmark feature of hypertension. However, whether maternal exercise during pregnancy has a sustained beneficial effect on the offspring of spontaneously hypertensive rats (SHRs) through epigenetic regulation of CaV1.2 channel is largely unknown. METHODS Pregnant SHRs and Wistar-Kyoto rats were subjected to swimming and the vascular molecular and functional properties of male offspring were evaluated at embryonic (E) 20.5 day, 3 months (3 M), and 6 months (6 M). RESULTS Exercise during pregnancy significantly decreased the resting blood pressure at 3 M but not 6 M in the offspring of SHR. Prenatal exercise significantly reduced the cardiovascular reactivity, the contribution of CaV1.2 channel to the vascular tone, and the whole-cell current density of CaV1.2 channel in both 3 M and 6 M offspring of SHR. Moreover, maternal exercise triggered hypermethylation of the promoter region of the CaV1.2 α1C gene (CACNA1C), with a concomitant decrease in its protein and mRNA expressions in SHR offspring at E20.5, 3 M, and 6 M. Tissue culture experiments further confirmed that 5-Aza-2'-deoxycytidine increased the structure and functional expression of CaV1.2 channel by inhibiting the DNA methylation of CACNA1C. However, the improvement of prenatal exercise on the blood pressure, function, and expression of CaV1.2 channel was attenuated in the offspring of SHRs at 6 M compared to the 3 M readout. CONCLUSIONS These data suggest that prenatal exercise improves the vascular function by the hypermethylation of CACNA1C in the arterial myocytes and delays the development of hypertension in the offspring of SHRs. However, these effects fade out with age.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; Department of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Yu Chen
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Yanyan Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Fang Qiu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Fanxing Zeng
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China.
| |
Collapse
|
19
|
Spiroski AM, Niu Y, Nicholas LM, Austin-Williams S, Camm EJ, Sutherland MR, Ashmore TJ, Skeffington KL, Logan A, Ozanne SE, Murphy MP, Giussani DA. Mitochondria antioxidant protection against cardiovascular dysfunction programmed by early-onset gestational hypoxia. FASEB J 2021; 35:e21446. [PMID: 33788974 PMCID: PMC7612077 DOI: 10.1096/fj.202002705r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023]
Abstract
Mitochondria-derived oxidative stress during fetal development increases cardiovascular risk in adult offspring of pregnancies complicated by chronic fetal hypoxia. We investigated the efficacy of the mitochondria-targeted antioxidant MitoQ in preventing cardiovascular dysfunction in adult rat offspring exposed to gestational hypoxia, integrating functional experiments in vivo, with those at the isolated organ and molecular levels. Rats were randomized to normoxic or hypoxic (13%-14% O2 ) pregnancy ± MitoQ (500 μM day-1 ) in the maternal drinking water. At 4 months of age, one cohort of male offspring was chronically instrumented with vascular catheters and flow probes to test in vivo cardiovascular function. In a second cohort, the heart was isolated and mounted onto a Langendorff preparation. To establish mechanisms linking gestational hypoxia with cardiovascular dysfunction and protection by MitoQ, we quantified the expression of antioxidant system, β-adrenergic signaling, and calcium handling genes in the fetus and adult, in frozen tissues from a third cohort. Maternal MitoQ in hypoxic pregnancy protected offspring against increased α1 -adrenergic reactivity of the cardiovascular system, enhanced reactive hyperemia in peripheral vascular beds, and sympathetic dominance, hypercontractility and diastolic dysfunction in the heart. Inhibition of Nfe2l2-mediated oxidative stress in the fetal heart and preservation of calcium regulatory responses in the hearts of fetal and adult offspring link molecular mechanisms to the protective actions of MitoQ treatment of hypoxic pregnancy. Therefore, these data show the efficacy of MitoQ in buffering mitochondrial stress through NADPH-induced oxidative damage and the prevention of programmed cardiovascular disease in adult offspring of hypoxic pregnancy.
Collapse
Affiliation(s)
- Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Lisa M Nicholas
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Shani Austin-Williams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Megan R Sutherland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Katie L Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Angela Logan
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Susan E Ozanne
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
20
|
Hula N, Spaans F, Vu J, Quon A, Kirschenman R, Cooke CLM, Phillips TJ, Case CP, Davidge ST. Placental treatment improves cardiac tolerance to ischemia/reperfusion insult in adult male and female offspring exposed to prenatal hypoxia. Pharmacol Res 2021; 165:105461. [PMID: 33513355 DOI: 10.1016/j.phrs.2021.105461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/08/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Offspring born from complicated pregnancies are at greater risk of cardiovascular disease in adulthood. Prenatal hypoxia is a common pregnancy complication that results in placental oxidative stress and impairs fetal development. Adult offspring exposed to hypoxia during fetal life are more susceptible to develop cardiac dysfunction, and show decreased cardiac tolerance to an ischemia/reperfusion (I/R) insult. To improve offspring cardiac outcomes, we have assessed the use of a placenta-targeted intervention during hypoxic pregnancies, by encapsulating the mitochondrial antioxidant MitoQ into nanoparticles (nMitoQ). We hypothesized that maternal nMitoQ treatment during hypoxic pregnancies improves cardiac tolerance to I/R insult in adult male and female offspring. Pregnant Sprague-Dawley rats were exposed to normoxia (21 % O2) or hypoxia (11 % O2) from gestational day 15-20, after injection with 100 μL saline or nMitoQ (125 μM) on GD15 (n=6-8/group). Male and female offspring were aged to 4 months. Both male and female offspring from hypoxic pregnancies showed reduced cardiac tolerance to I/R (assessed ex vivo using the isolated working heart technique) which was ameliorated by nMitoQ treatment. To identify potential molecular mechanisms for the changes in cardiac tolerance to I/R, cardiac levels/phosphorylation of proteins important for intracellular Ca2+ cycling were assessed with Western blotting. In prenatally hypoxic male offspring, improved cardiac recovery from I/R by nMitoQ was accompanied by increased cardiac phospholamban and phosphatase 2Ce levels, and a trend to decreased Ca2+/calmodulin-dependent protein kinase IIδ phosphorylation. In contrast, in female offspring, nMitoQ treatment in hypoxic pregnancies increased phospholamban and protein kinase Cε phosphorylation. Maternal nMitoQ treatment improves cardiac tolerance to I/R insult in adult offspring and thus has the potential to improve the later-life trajectory of cardiovascular health of adult offspring born from pregnancies complicated by prenatal hypoxia.
Collapse
Affiliation(s)
- Nataliia Hula
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Jennie Vu
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Tom J Phillips
- Dementia Research Institute, Cardiff University, Cardiff, CF10 3AT, UK.
| | - C Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol, BS10 5NB, UK.
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| |
Collapse
|
21
|
Spiroski AM, Sanders R, Meloni M, McCracken IR, Thomson A, Brittan M, Gray GA, Baker AH. The Influence of the LINC00961/SPAAR Locus Loss on Murine Development, Myocardial Dynamics, and Cardiac Response to Myocardial Infarction. Int J Mol Sci 2021; 22:ijms22020969. [PMID: 33478078 PMCID: PMC7835744 DOI: 10.3390/ijms22020969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 01/14/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have structural and functional roles in development and disease. We have previously shown that the LINC00961/SPAAR (small regulatory polypeptide of amino acid response) locus regulates endothelial cell function, and that both the lncRNA and micropeptide counter-regulate angiogenesis. To assess human cardiac cell SPAAR expression, we mined a publicly available scRNSeq dataset and confirmed LINC00961 locus expression and hypoxic response in a murine endothelial cell line. We investigated post-natal growth and development, basal cardiac function, the cardiac functional response, and tissue-specific response to myocardial infarction. To investigate the influence of the LINC00961/SPAAR locus on longitudinal growth, cardiac function, and response to myocardial infarction, we used a novel CRISPR/Cas9 locus knockout mouse line. Data mining suggested that SPAAR is predominantly expressed in human cardiac endothelial cells and fibroblasts, while murine LINC00961 expression is hypoxia-responsive in mouse endothelial cells. LINC00961–/– mice displayed a sex-specific delay in longitudinal growth and development, smaller left ventricular systolic and diastolic areas and volumes, and greater risk area following myocardial infarction compared with wildtype littermates. These data suggest the LINC00961/SPAAR locus contributes to cardiac endothelial cell and fibroblast function and hypoxic response, growth and development, and basal cardiovascular function in adulthood.
Collapse
Affiliation(s)
- Ana-Mishel Spiroski
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Rachel Sanders
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Marco Meloni
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Ian R. McCracken
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Adrian Thomson
- Edinburgh Preclinical Imaging, Edinburgh Preclinical Imaging, BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Mairi Brittan
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Gillian A. Gray
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
| | - Andrew H. Baker
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (A.-M.S.); (R.S.); (M.M.); (I.R.M.); (M.B.); (G.A.G.)
- Correspondence: ; Tel.: +44-0131-24-26728
| |
Collapse
|
22
|
Hellgren KT, Premanandhan H, Quinn CJ, Trafford AW, Galli GLJ. Sex-dependent effects of developmental hypoxia on cardiac mitochondria from adult murine offspring. Free Radic Biol Med 2021; 162:490-499. [PMID: 33186741 DOI: 10.1016/j.freeradbiomed.2020.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Insufficient oxygen supply (hypoxia) during fetal and embryonic development can lead to latent phenotypical changes in the adult cardiovascular system, including altered cardiac function and increased susceptibility to ischemia reperfusion injury. While the cellular mechanisms underlying this phenomenon are largely unknown, several studies have pointed towards metabolic disturbances in the heart of offspring from hypoxic pregnancies. To this end, we investigated mitochondrial function in the offspring of a mouse model of prenatal hypoxia. Pregnant C57 mice were subjected to either normoxia (21%) or hypoxia (14%) during gestational days 6-18. Offspring were reared in normoxia for up to 8 months and mitochondrial biology was assessed with electron microscopy (ultrastructure), spectrophotometry (enzymatic activity of electron transport chain complexes), microrespirometry (oxidative phosphorylation and H202 production) and Western Blot (protein expression). Our data showed that male adult offspring from hypoxic pregnancies possessed mitochondria with increased H202 production and lower respiratory capacity that was associated with reduced protein expression of complex I, II and IV. In contrast, females from hypoxic pregnancies had a higher respiratory capacity and lower H202 production that was associated with increased enzymatic activity of complex IV. From these results, we speculate that early exposure to hypoxia has long term, sex-dependent effects on cardiac metabolic function, which may have implications for cardiovascular health and disease in adulthood.
Collapse
Affiliation(s)
- Kim T Hellgren
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Hajani Premanandhan
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Callum J Quinn
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Gina L J Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK.
| |
Collapse
|
23
|
Yan Z, Liqiong S, Yingduo Y, Jin Q, Boyang Y. Application of multi-dimensional and multi-informational (MD-MI) integrated xanthine oxidase and superoxide anion fingerprint in quality evaluation of Scutellariae Radix. J Pharm Biomed Anal 2020; 191:113595. [PMID: 32905858 DOI: 10.1016/j.jpba.2020.113595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/02/2020] [Accepted: 08/23/2020] [Indexed: 10/23/2022]
Abstract
A multi-hyphenated analytical method that was successfully established in previous research was applied to quality evaluation of traditional Chinese medicine (TCM) to verify its feasibility in complex systems. Scutellariae Radix (SR), which significantly protects against oxidative damage from ischemia and reperfusion, was selected as the TCM for this study. A dual-activity detection system based on xanthine oxidase (XOD) inhibition and superoxide anion (O2-) scavenging activity was used to generate a multi-dimensional-multi-informational (MD-MI) integrated fingerprint of SR. Combined with HPLC-ESI-Q-TOF-MS analysis, 17 active compounds in SR were tentatively identified by comparison with reference substances or literature data. The quality of SR from different habitats was comprehensively and systematically evaluated in respect of chemical composition, XOD inhibition and O2- scavenging activity. It was confirmed that SR contains many antioxidants and XOD inhibitory substances with diverse functions. Among them, baicalin, norwogonin-7-O-glucuronide and baicalein are the main contributors to direct antioxidant activity. Acteoside, 5,7,2',5'-tetrahydroxy-8,6'-dimethoxy flavone, baicalin and baicalein are the main XOD inhibitory components of SR. Comprehensive analysis found that the antioxidant activity of SR from Gansu Province was superior to that from other provinces in terms of both XOD inhibition and O2- scavenging activity. It has been demonstrated that the method is capable of analyzing complex TCM matrices, and can provide a useful reference for establishing quality control of TCM from the perspective of MD-MI.
Collapse
Affiliation(s)
- Zhu Yan
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Sun Liqiong
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing 210095, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yang Yingduo
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Qi Jin
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Yu Boyang
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
24
|
Hess RM, Niu Y, Garrud TAC, Botting KJ, Ford SG, Giussani DA. Embryonic cardioprotection by hydrogen sulphide: studies of isolated cardiac function and ischaemia-reperfusion injury in the chicken embryo. J Physiol 2020; 598:4197-4208. [PMID: 32705691 DOI: 10.1113/jp279978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/22/2020] [Indexed: 01/06/2023] Open
Abstract
KEY POINTS In mammals, pregnancy complications can trigger an embryonic or fetal origin of cardiac dysfunction. However, underlying mechanisms remain uncertain because the partial contributions of the challenge on the mother, placenta or offspring are difficult to disentangle. The avian embryo permits isolation of the direct effects of suboptimal conditions during development on the cardiac function of the offspring, independent of additional effects on the mother and/or the placenta. Therefore, the objectives of this work were to adapt the isolated Langendorff technique using the chicken embryo to study the physiology of the developing heart. Here, we introduce a novel technique and show the utility of the technique for exploring cardioprotective roles of H2 S in the chicken embryo heart. This work lays the foundation for studying the direct effects of H2 S therapy on the embryonic heart independent of effects on the mother and the placenta in adverse development. ABSTRACT This study adapted the isolated Langendorff preparation to study the chicken embryo heart in response to ischaemia-reperfusion (IR) injury. The utility of the technique was tested by investigating cardioprotective effects of hydrogen sulphide (H2 S) and underlying mechanisms. Embryonic hearts (19 out of 21 days of incubation) mounted on a Langendorff preparation were exposed to IR (30 min ischaemia) after 4 treatments administered randomly, all as a 1 mm bolus, into the perfusate: saline vehicle (control); sodium hydrogen sulphide (NaHS); NaHS plus glibenclamide, an antagonist of KATP opening (NaHS Glib), and Glib alone (Glib). Relative to controls, NaHS treatment improved cardiac function after ischaemia (mean ± SD for area under the curve, AUC, for left ventricular developed pressure, LVDP: 1767.3 ± 929.5 vs. 492.7 ± 308.1; myocardial contractility, dP/dtmax : 2748.9 ± 1514.9 vs. 763.7 ± 433.1) and decreased infarct size (22.7 ± 8.0 vs. 43.9 ± 4.2%) and cardiac damage (% change in creatinine kinase, 49.3 ± 41.3 vs. 214.6 ± 155.1; all P < 0.05). Beneficial effects of NaHS were blocked by Glib. Glib alone had no effects. NaHS increased coronary flow rate (CFR) during baseline (mean ± SD for AUC: 134.3 ± 91.6 vs. 92.2 ± 35.8) and post IR (1467 ± 529.5 vs. 748.0 ± 222.1; both P < 0.05). However, this effect was not prevented by Glib. Therefore, the chicken embryo heart is amenable for study via the Langendorff preparation under basal conditions and during IR. The data show that H2 S confers embryonic cardiac protection via opening of myocardial KATP channels and not via increasing CFR. H2 S may prove a useful therapeutic agent to protect the human fetal heart against IR injury, as may occur in complicated labour.
Collapse
Affiliation(s)
- Rita M Hess
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiativ, University of Cambridge, UK
| | - Tessa A C Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiativ, University of Cambridge, UK
| | - Sage G Ford
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiativ, University of Cambridge, UK.,Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, UK
| |
Collapse
|
25
|
Skeffington KL, Beck C, Itani N, Niu Y, Shaw CJ, Giussani DA. Hypertension Programmed in Adult Hens by Isolated Effects of Developmental Hypoxia In Ovo. Hypertension 2020; 76:533-544. [PMID: 32536277 PMCID: PMC7340221 DOI: 10.1161/hypertensionaha.120.15045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In mammals, pregnancy complicated by chronic hypoxia can program hypertension in the adult offspring. However, mechanisms remain uncertain because the partial contributions of the challenge on the placenta, mother, and fetus are difficult to disentangle. Here, we used chronic hypoxia in the chicken embryo-an established model system that permits isolation of the direct effects of developmental hypoxia on the cardiovascular system of the offspring, independent of additional effects on the mother or the placenta. Fertilized chicken eggs were exposed to normoxia (N; 21% O2) or hypoxia (H; 13.5%-14% O2) from the start of incubation (day 0) until day 19 (hatching, ≈day 21). Following hatching, all birds were maintained under normoxic conditions until ≈6 months of adulthood. Hypoxic incubation increased hematocrit (+27%) in the chicken embryo and induced asymmetrical growth restriction (body weight, -8.6%; biparietal diameter/body weight ratio, +7.5%) in the hatchlings (all P<0.05). At adulthood (181±4 days), chickens from hypoxic incubations remained smaller (body weight, -7.5%) and showed reduced basal and stimulated in vivo NO bioavailability (pressor response to NG-nitro-L-arginine methyl ester, -43%; phenylephrine pressor response during NO blockade, -61%) with significant hypertension (mean arterial blood pressure, +18%), increased cardiac work (ejection fraction, +12%; fractional shortening, +25%; enhanced baroreflex gain, +456%), and left ventricular wall thickening (left ventricular wall volume, +36%; all P<0.05). Therefore, we show that chronic hypoxia can act directly on a developing embryo to program hypertension, cardiovascular dysfunction, and cardiac wall remodeling in adulthood in the absence of any maternal or placental effects.
Collapse
Affiliation(s)
- Katie L. Skeffington
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Christian Beck
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Nozomi Itani
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Youguo Niu
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Caroline J. Shaw
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.),Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, United Kingdom (C.J.S.)
| | - Dino A. Giussani
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| |
Collapse
|
26
|
Ganguly E, Hula N, Spaans F, Cooke CLM, Davidge ST. Placenta-targeted treatment strategies: An opportunity to impact fetal development and improve offspring health later in life. Pharmacol Res 2020; 157:104836. [PMID: 32344051 DOI: 10.1016/j.phrs.2020.104836] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/26/2020] [Accepted: 04/13/2020] [Indexed: 01/06/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) theory states that a sub-optimal prenatal and early postnatal environment during development leads to an increased risk of long-term development of adult chronic diseases. Developmental programming of disease has the potential to greatly impact the health of our population. Therefore, research has focused on the development of primary treatment strategies and/or therapeutic interventions for individuals who are at increased risk, with the objective to reverse or prevent later life onset of chronic disease in the offspring born from complicated pregnancies. Many studies have focused on systemic treatments and/or interventions in complicated pregnancies to improve offspring outcomes. However, there are limitations to systemic maternal/prenatal treatments, as most of the treatments are able to cross the placenta and have potential adverse off-target effects on the developing fetus. The placenta serves as the primary interface between mother and fetus, and placental dysfunction in complicated pregnancies has been associated with impaired fetal development and negative impact on offspring health. Therefore, recent research has focused on treatment strategies that specifically target the placenta to improve placental function and prevent passage of prenatal therapeutics and/or treatments into the fetal circulation, thus avoiding any potential adverse off-target effects on the fetus. This article reviews the currently available knowledge on treatment strategies and/or therapeutics that specifically target the placenta with the goal of improving pregnancy outcomes with a focus on long-term health of the offspring born of complicated pregnancies.
Collapse
Affiliation(s)
- Esha Ganguly
- Department of Physiology, University of Alberta, Edmonton, Canada; Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Nataliia Hula
- Department of Physiology, University of Alberta, Edmonton, Canada; Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, Canada; Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
27
|
Gatford KL, Andraweera PH, Roberts CT, Care AS. Animal Models of Preeclampsia: Causes, Consequences, and Interventions. Hypertension 2020; 75:1363-1381. [PMID: 32248704 DOI: 10.1161/hypertensionaha.119.14598] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preeclampsia is a common pregnancy complication, affecting 2% to 8% of pregnancies worldwide, and is an important cause of both maternal and fetal morbidity and mortality. Importantly, although aspirin and calcium are able to prevent preeclampsia in some women, there is no cure apart from delivery of the placenta and fetus, often necessitating iatrogenic preterm birth. Preclinical models of preeclampsia are widely used to investigate the causes and consequences of preeclampsia and to evaluate safety and efficacy of potential preventative and therapeutic interventions. In this review, we provide a summary of the published preclinical models of preeclampsia that meet human diagnostic criteria, including the development of maternal hypertension, together with new-onset proteinuria, maternal organ dysfunction, and uteroplacental dysfunction. We then discuss evidence from preclinical models for multiple causal factors of preeclampsia, including those implicated in early-onset and late-onset preeclampsia. Next, we discuss the impact of exposure to a preeclampsia-like environment for later maternal and progeny health. The presence of long-term impairment, particularly cardiovascular outcomes, in mothers and progeny after an experimentally induced preeclampsia-like pregnancy, implies that later onset or reduced severity of preeclampsia will improve later maternal and progeny health. Finally, we summarize published intervention studies in preclinical models and identify gaps in knowledge that we consider should be targets for future research.
Collapse
Affiliation(s)
- Kathryn L Gatford
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Prabha H Andraweera
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Claire T Roberts
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Alison S Care
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| |
Collapse
|
28
|
Frasch MG, Herry CL, Niu Y, Giussani DA. First evidence that intrinsic fetal heart rate variability exists and is affected by hypoxic pregnancy. J Physiol 2020; 598:249-263. [PMID: 31802494 DOI: 10.1113/jp278773] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/11/2019] [Indexed: 02/02/2023] Open
Abstract
KEY POINTS We introduce a technique to test whether intrinsic fetal heart rate variability (iFHRV) exists and we show the utility of the technique by testing the hypothesis that iFHRV is affected by chronic fetal hypoxia, one of the most common adverse outcomes of human pregnancy complicated by fetal growth restriction. Using an established late gestation ovine model of fetal development under chronic hypoxic conditions, we identify iFHRV in isolated fetal hearts and show that it is markedly affected by hypoxic pregnancy. Therefore, the isolated fetal heart has intrinsic variability and carries a memory of adverse intrauterine conditions experienced during the last third of pregnancy. ABSTRACT Fetal heart rate variability (FHRV) emerges from influences of the autonomic nervous system, fetal body and breathing movements, and from baroreflex and circadian processes. We tested whether intrinsic heart rate variability (iHRV), devoid of any external influences, exists in the fetal period and whether it is affected by chronic fetal hypoxia. Chronically catheterized ewes carrying male singleton fetuses were exposed to normoxia (n = 6) or hypoxia (10% inspired O2 , n = 9) for the last third of gestation (105-138 days of gestation (dG); term ∼145 dG) in isobaric chambers. At 138 dG, isolated hearts were studied using a Langendorff preparation. We calculated basal intrinsic FHRV (iFHRV) indices reflecting iFHRV's variability, predictability, temporal symmetry, fractality and chaotic behaviour, from the systolic peaks within 15 min segments in each heart. Significance was assumed at P < 0.05. Hearts of fetuses isolated from hypoxic pregnancy showed approximately 4-fold increases in the Grid transformation as well as the AND similarity index (sgridAND) and a 4-fold reduction in the scale-dependent Lyapunov exponent slope. We also detected a 2-fold reduction in the Recurrence quantification analysis, percentage of laminarity (pL) and recurrences, maximum and average diagonal line (dlmax, dlmean) and the Multiscale time irreversibility asymmetry index. The iHRV measures dlmax, dlmean, pL and sgridAND correlated with left ventricular end-diastolic pressure across both groups (average R2 = 0.38 ± 0.03). This is the first evidence that iHRV originates in fetal life and that chronic fetal hypoxia significantly alters it. Isolated fetal hearts from hypoxic pregnancy exhibit a time scale-dependent higher complexity in iFHRV.
Collapse
Affiliation(s)
- Martin G Frasch
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Christophe L Herry
- Dynamical Analysis Laboratory, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Youguo Niu
- Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Nathanielsz PW. Maternal programming by hypoxia alters the molecular composition of the oviduct of her offspring, the first pathway her grandchildren will transit: a potential novel pathway for intergenerational programming? J Physiol 2019; 597:2325-2326. [PMID: 30883756 PMCID: PMC6487917 DOI: 10.1113/jp277868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
30
|
Brain KL, Allison BJ, Niu Y, Cross CM, Itani N, Kane AD, Herrera EA, Skeffington KL, Botting KJ, Giussani DA. Intervention against hypertension in the next generation programmed by developmental hypoxia. PLoS Biol 2019; 17:e2006552. [PMID: 30668572 PMCID: PMC6342530 DOI: 10.1371/journal.pbio.2006552] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Evidence derived from human clinical studies and experimental animal models shows a causal relationship between adverse pregnancy and increased cardiovascular disease in the adult offspring. However, translational studies isolating mechanisms to design intervention are lacking. Sheep and humans share similar precocial developmental milestones in cardiovascular anatomy and physiology. We tested the hypothesis in sheep that maternal treatment with antioxidants protects against fetal growth restriction and programmed hypertension in adulthood in gestation complicated by chronic fetal hypoxia, the most common adverse consequence in human pregnancy. Using bespoke isobaric chambers, chronically catheterized sheep carrying singletons underwent normoxia or hypoxia (10% oxygen [O2]) ± vitamin C treatment (maternal 200 mg.kg-1 IV daily) for the last third of gestation. In one cohort, the maternal arterial blood gas status, the value at which 50% of the maternal hemoglobin is saturated with oxygen (P50), nitric oxide (NO) bioavailability, oxidative stress, and antioxidant capacity were determined. In another, naturally delivered offspring were raised under normoxia until early adulthood (9 months). Lambs were chronically instrumented and cardiovascular function tested in vivo. Following euthanasia, femoral arterial segments were isolated and endothelial function determined by wire myography. Hypoxic pregnancy induced fetal growth restriction and fetal oxidative stress. At adulthood, it programmed hypertension by enhancing vasoconstrictor reactivity and impairing NO-independent endothelial function. Maternal vitamin C in hypoxic pregnancy improved transplacental oxygenation and enhanced fetal antioxidant capacity while increasing NO bioavailability, offsetting constrictor hyper-reactivity and replenishing endothelial function in the adult offspring. These discoveries provide novel insight into mechanisms and interventions against fetal growth restriction and adult-onset programmed hypertension in an animal model of complicated pregnancy in a species of similar temporal developmental milestones to humans.
Collapse
Affiliation(s)
- Kirsty L. Brain
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Beth J. Allison
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Youguo Niu
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, United Kingdom
| | - Christine M. Cross
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Nozomi Itani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andrew D. Kane
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Emilio A. Herrera
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Katie L. Skeffington
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley J. Botting
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, United Kingdom
| | - Dino A. Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|