1
|
Putra M, Peek EEH, Devore GR, Hobbins JC. Umbilical Vein Flows and Cardiac Size, Shape, and Ventricular Contractility in Fetuses With Estimated Weight Less-Than 10th Centile. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:2069-2084. [PMID: 39076048 DOI: 10.1002/jum.16536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/23/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVES In a cohort of patients with estimated fetal weights (EFWs) <10th centile, we aimed 1) to compare the prevalence of abnormalities of fetal 4-chamber view (4CV) cardiac size, shape, and ventricular contractility in fetal growth restricted (FGR) and small-for-gestational-age (SGA) fetuses and 2) to compare umbilical vein flow (UVF) measurements to standard Doppler surveillance in predicting abnormalities of cardiac function. METHODS Prospective observational cohort study of fetuses with EFW <10th percentile. Measurements of size and shape used were 4CV transverse width, 4CV cardiac area, 4CV global sphericity index, and right-to-left ventricular mid-chamber width ratio. Variables of contractility used were fractional shortening change at the mid-ventricle chamber, global longitudinal strain, fractional area change, and left ventricular cardiac output. The UVF and standard Doppler surveillance including umbilical artery (UA), middle cerebral artery, and cerebroplacental ratio (CPR) were collected. Control data were from previously published studies. RESULTS A total of 95 fetuses with EFWs <10th centile were included in the study. The rates of abnormalities of cardiac size and shape and ventricular contractility were all significantly elevated compared with normally grown control fetuses but similar between FGR and SGA fetuses. In a subset of 76 patients with UVF data, evaluation UVF identified more patients with any abnormality of contractility compared with UA (37.9 vs 17.2%, P = .02). CONCLUSIONS The addition of UVF doubled the detection rate of ventricular contractility abnormalities. The addition of UVF should be considered in the surveillance of FGR and SGA fetuses to further stratify the severity of hypoxemia and to identify those at greater risk for future cardiovascular dysfunction.
Collapse
Affiliation(s)
- Manesha Putra
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Greggory R Devore
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
- Fetal Diagnostic Centers, Lancaster, California, USA
| | - John C Hobbins
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
2
|
Heath-Freudenthal A, Estrada A, von Alvensleben I, Julian CG. Surviving birth at high altitude. J Physiol 2024; 602:5463-5473. [PMID: 38520695 PMCID: PMC11418585 DOI: 10.1113/jp284554] [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: 07/04/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
This Symposium Review examines challenges to surviving birth and infancy at high altitudes. Chronic exposure to the environmental hypoxia of high altitudes increases the incidence of maternal vascular disorders of pregnancy characterized by placental insufficiency, restricted fetal growth and preterm delivery, and impairs pulmonary vascular health during infancy. While each condition independently contributes to excess morbidity and mortality in early life, evidence indicates vascular disorders of pregnancy and infantile pulmonary vascular dysfunction are intertwined. By integrating our recent scientific and clinical observations in Bolivia with existing literature, we propose potential avenues to reduce the infant mortality burden at high altitudes and reduce pulmonary vascular disease in highland neonates, and emphasize the need for further research to address unresolved questions.
Collapse
Affiliation(s)
| | | | | | - Colleen G. Julian
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, US
| |
Collapse
|
3
|
Rashidi SY, Rafiyan M, Asemi R, Asemi Z, Mohammadi S. Effect of melatonin as a therapeutic strategy against intrauterine growth restriction: a mini-review of current state. Ann Med Surg (Lond) 2024; 86:5320-5325. [PMID: 39238981 PMCID: PMC11374193 DOI: 10.1097/ms9.0000000000002350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
Intrauterine growth restriction (IUGR) or intrauterine growth retardation is a condition that the fetus does not grow as expected. And the biometric profile does not match with the age of fetus. This condition is associated with increased mortality and morbidity of the neonates along with increased risk of cardiovascular, lung, and central nervous system damage. Despite close monitoring of high-risk mothers and the development of new therapeutic approaches, the optimal outcome has not been achieved yet that it indicates the importance of investigations on new therapeutic approaches. Melatonin (MLT) is a neurohormone mainly produced by the pineal gland and has a wide range of effects on different organs due to the broad dispersion of its receptors. Moreover, melatonin is produced by the placenta and also its receptors have been found on the surface of this organ. Not only studies showed the importance of this neurohormone on growth and development of fetus but also they proved its highly anti-oxidant properties. As in IUGR the oxidative stress and inflammation increased melatonin could counteract these changes and improved organ's function. In this study, we found that use of MLT could be a good clinical approach for the treatment of IUGR as its high anti-oxidant activity and vasodilation could dampen the mechanisms lead to the IUGR development.
Collapse
Affiliation(s)
| | - Mahdi Rafiyan
- Student Research Committee
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan
| | - Sotoudeh Mohammadi
- Department of Obstetrics and Gynecology, Shahid Beheshti university of medical sciences, Tehran, Iran
| |
Collapse
|
4
|
van de Meent M, Nijholt KT, Joemmanbaks SCA, Kooiman J, Schipper HS, Wever KE, Lely AT, Terstappen F. Understanding changes in echocardiographic parameters at different ages following fetal growth restriction: a systematic review and meta-analysis. Am J Physiol Heart Circ Physiol 2024; 326:H1469-H1488. [PMID: 38668703 PMCID: PMC11380958 DOI: 10.1152/ajpheart.00052.2024] [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: 01/30/2024] [Revised: 03/29/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fetal growth restriction (FGR) increases cardiovascular risk by cardiac remodeling and programming. This systematic review and meta-analysis across species examines the use of echocardiography in FGR offspring at different ages. PubMed and Embase.com were searched for animal and human studies reporting on echocardiographic parameters in placental insufficiency-induced FGR offspring. We included six animal and 49 human studies. Although unable to perform a meta-analysis of animal studies because of insufficient number of studies per individual outcome, all studies showed left ventricular dysfunction. Our meta-analyses of human studies revealed a reduced left ventricular mass, interventricular septum thickness, mitral annular peak velocity, and mitral lateral early diastolic velocity at neonatal age. No echocardiographic differences during childhood were observed, although the small age range and number of studies limited these analyses. Only two studies at adult age were performed. Meta-regression on other influential factors was not possible due to underreporting. The few studies on myocardial strain analysis showed small changes in global longitudinal strain in FGR offspring. The quality of the human studies was considered low and the risk of bias in animal studies was mostly unclear. Echocardiography may offer a noninvasive tool to detect early signs of cardiovascular predisposition following FGR. Clinical implementation yet faces multiple challenges including identification of the most optimal timing and the exact relation to long-term cardiovascular function in which echocardiography alone might be limited to reflect a child's vascular status. Future research should focus on myocardial strain analysis and the combination of other (non)imaging techniques for an improved risk estimation.NEW & NOTEWORTHY Our meta-analysis revealed echocardiographic differences between fetal growth-restricted and control offspring in humans during the neonatal period: a reduced left ventricular mass and interventricular septum thickness, reduced mitral annular peak velocity, and mitral lateral early diastolic velocity. We were unable to pool echocardiographic parameters in animal studies and human adults because of an insufficient number of studies per individual outcome. The few studies on myocardial strain analysis showed small preclinical changes in FGR offspring.
Collapse
Affiliation(s)
- Mette van de Meent
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Kirsten T Nijholt
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Shary C A Joemmanbaks
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Judith Kooiman
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Henk S Schipper
- Department of Pediatric Cardiology, Sophia Children's Hospital, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kimberley E Wever
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A Titia Lely
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Fieke Terstappen
- Division Women and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
- Division Women and Baby, Department of Neonatology, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Ye L, Huang Y, Chen K, Hang C, Ying Y, Zu L, Luo X, Du L. Early postnatal moderate catch‑up growth in rats with nutritional intrauterine growth restriction preserves pulmonary vascular and cognitive function in adulthood. Exp Ther Med 2024; 27:183. [PMID: 38515647 PMCID: PMC10952380 DOI: 10.3892/etm.2024.12471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
Intrauterine growth restriction (IUGR) with rapid postnatal catch-up growth is strongly associated with pulmonary vascular dysfunction in adulthood, whereas IUGR with delayed growth in early postnatal life results in long-term brain deficits. In the present study, it was hypothesized that IUGR with early moderate catch-up growth may alleviate pulmonary vascular remodeling in adulthood without affecting memory function. An IUGR model was established by restricting maternal nutrition during pregnancy. Different growth patterns were achieved by adjusting the litter size in each group during lactation. Rats meeting the weight requirement at weaning were selected for subsequent studies at three time points (3, 9 and 13 weeks). Cognitive function was evaluated using a Y-maze. Invasive hemodynamic measurements were conducted to measure the mean pulmonary arterial pressure (mPAP). In addition, primary pulmonary artery smooth muscle cells (PASMCs) and pulmonary vascular endothelial cells (PVECs) were cultured to investigate their role in the increase in mPAP following rapid catch-up growth. The results showed that memory function deficits in the rats in the delayed growth group were associated with reduced proliferation of neural stem cells in the subgranular zone of the hippocampus. Furthermore, moderate catch-up growth at the three time points improved memory function while maintaining a normal mPAP. In adult IUGR rats experiencing rapid catch-up growth, although memory function improved, elevated mPAP and medial thickening of pulmonary arterioles were observed. Additionally, PASMCs exhibited excessive proliferation, migration and anti-apoptotic activity in the rapid catch-up group, and PVECs also displayed excessive proliferation. These results suggested that moderate catch-up growth after IUGR is a better strategy for optimal cognition and cardiovascular health in adulthood compared with rapid catch-up growth or delayed growth.
Collapse
Affiliation(s)
- Lixia Ye
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Yajie Huang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Chengcheng Hang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Yuhan Ying
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Lu Zu
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Xiaofei Luo
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Lizhong Du
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| |
Collapse
|
6
|
Wooldridge AL, Kirschenman R, Spaans F, Pasha M, Davidge ST, Cooke CLM. Advanced maternal age alters cardiac functional and structural adaptations to pregnancy in rats. Am J Physiol Heart Circ Physiol 2024; 326:H1131-H1137. [PMID: 38456848 DOI: 10.1152/ajpheart.00057.2024] [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: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
A significant number of pregnancies occur at advanced maternal age (>35 yr), which is a risk factor for pregnancy complications. Healthy pregnancies require massive hemodynamic adaptations, including an increased blood volume and cardiac output. There is growing evidence that these cardiovascular adaptations are impaired with age, however, little is known about maternal cardiac function with advanced age. We hypothesized that cardiac adaptations to pregnancy are impaired with advanced maternal age. Younger (4 mo; ∼early reproductive maturity in humans) and aged (9 mo; ∼35 yr in humans) pregnant Sprague-Dawley rats were assessed and compared with age-matched nonpregnant controls. Two-dimensional echocardiographic images were obtained (ultrasound biomicroscopy; under anesthesia) on gestational day 19 (term = 22 days) and compared with age-matched nonpregnant rats (n = 7-9/group). Left ventricular structure and function were assessed using short-axis images and transmitral Doppler signals. During systole, left ventricular anterior wall thickness increased with age in the nonpregnant rats, but there was no age-related difference between the pregnant groups. There were no significant pregnancy-associated differences in left ventricular wall thickness. Calculated left ventricular mass increased with age in nonpregnant rats and increased with pregnancy only in young rats. Compared with young pregnant rats, the aortic ejection time of aged pregnant rats was greater and Tei index was lower. Overall, the greater aortic ejection time and lower Tei index with age in pregnant rats suggest mildly altered cardiac adaptations to pregnancy with advanced maternal age, which may contribute to adverse outcomes in advanced maternal age pregnancies.NEW & NOTEWORTHY We demonstrated that even before the age of reproductive senescence, rats show signs of age-related alterations in cardiac structure that suggests increased cardiac work. Our data also demonstrate, using an in vivo echocardiographic approach, that advanced maternal age in a rat model is associated with altered cardiac function and structure relative to younger pregnant controls.
Collapse
Affiliation(s)
- Amy L Wooldridge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Mazhar Pasha
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Rodríguez-López M, Sepúlveda-Martínez Á, Bernardino G, Crovetto F, Pajuelo C, Sitges M, Bijnens B, Gratacós E, Crispi F. Cardiometabolic sex differences in adults born small for gestational age. Front Cardiovasc Med 2023; 10:1223928. [PMID: 37953765 PMCID: PMC10634502 DOI: 10.3389/fcvm.2023.1223928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023] Open
Abstract
Aim This study aimed to assess the cardiometabolic sex similarities and differences in adults born small for gestational age. Methods This study was an ambispective cohort study from a birth registry in Barcelona, Spain, including 523 adult participants (20-40 years-old) subdivided as born small for gestational age (SGA, if birth weight <10th centile) or adequate fetal growth for gestational age (AGA). Cardiometabolic health was assessed by echocardiography, electrocardiogram, blood pressure measurement, vascular ultrasound, anthropometric measurements, and serum glycemia and lipid profile. Stratified analyses by sex were performed by estimation of adjusted absolute difference (AAD) using inverse probability weighting. Results Compared with AGA, the stratified analyses by sex showed a more pronounced reduction in ejection fraction [AAD: female -1.73 (95% CI -3.2 to -0.28) vs. male -1.33 (-3.19 to 0.52)] and increment in heart rate [female 3.04 (0.29-5.8) vs. male 2.25 (-0.82 to 5.31)] in SGA females compared with SGA males. In contrast, a more pronounced reduction in PR interval [female -1.36 (-6.15 to 3.42) vs. male -6.61 (-11.67 to -1.54)] and an increase in systolic blood pressure [female 0.06 (-2.7 to 2.81) vs. male 2.71 (-0.48 to 5.9)] and central-to-peripheral fat ratio [female 0.05 (-0.03 to 0.12) vs. male 0.40 (0.17-0.62)] were mainly observed in SGA male compared with SGA female. Conclusions Sex differences were observed in the effect of SGA on cardiometabolic endpoints with female being more prone to cardiac dysfunction and male to electrocardiographic, vascular, and metabolic changes. Future research including sex-stratification data is warranted.
Collapse
Affiliation(s)
- Mérida Rodríguez-López
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Centro de Investigaciones Biomédica en Red – Enfermedades Raras, Universitat de Barcelona, Barcelona, Spain
- Faculty of Health Science, Universidad Icesi, Cali, Colombia
- Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
| | - Álvaro Sepúlveda-Martínez
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Centro de Investigaciones Biomédica en Red – Enfermedades Raras, Universitat de Barcelona, Barcelona, Spain
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Clínico Universidad de Chile, Santiago de Chile, Chile
| | - Gabriel Bernardino
- BCN Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Centro de Investigaciones Biomédica en Red – Enfermedades Raras, Universitat de Barcelona, Barcelona, Spain
| | - Carolina Pajuelo
- Institut Clínic Cardiovascular, Hospital Clínic, Centre for Biomedical Research on CardioVascular Diseases (CIBERCV), Universitat de Barcelona, Barcelona, Spain
| | - Marta Sitges
- Institut Clínic Cardiovascular, Hospital Clínic, Centre for Biomedical Research on CardioVascular Diseases (CIBERCV), Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
| | - Bart Bijnens
- Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Eduard Gratacós
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Centro de Investigaciones Biomédica en Red – Enfermedades Raras, Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
| | - Fàtima Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Centro de Investigaciones Biomédica en Red – Enfermedades Raras, Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
| |
Collapse
|
8
|
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
|
9
|
Whitlock AE, Moskowitzova K, Kycia I, Zurakowski D, Fauza DO. Morphometric, Developmental, and Anti-Inflammatory Effects of Transamniotic Stem Cell Therapy (TRASCET) on the Fetal Heart and Lungs in a Model of Intrauterine Growth Restriction. Stem Cells Dev 2023; 32:484-490. [PMID: 37358376 DOI: 10.1089/scd.2023.0040] [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] [Indexed: 06/27/2023] Open
Abstract
Transamniotic stem cell therapy (TRASCET) with mesenchymal stem cells (MSCs) can attenuate placental inflammation and minimize intrauterine growth restriction (IUGR). We sought to determine whether MSC-based TRASCET could mitigate fetal cardiopulmonary effects of IUGR. Pregnant Sprague-Dawley dams were exposed to alternating 12-h hypoxia (10.5% O2) cycles in the last fourth of gestation. Their fetuses (n = 155) were divided into 4 groups. One group remained untreated (n = 42), while three groups received volume-matched intra-amniotic injections of either saline (sham; n = 34), or of syngeneic amniotic fluid-derived MSCs, either in their native state (TRASCET; n = 36) or "primed" by exposure to interferon-gamma and interleukin-1beta before administration in vivo (TRASCET-primed; n = 43). Normal fetuses served as additional controls (n = 30). Multiple morphometric and biochemical analyses were performed at term for select markers of cardiopulmonary development and inflammation previously shown to be affected by IUGR. Among survivors (75%; 117/155), fetal heart-to-body weight ratio was increased in both the sham and untreated groups (P < 0.001 for both) but normalized in the TRASCET and TRASCET-primed groups (P = 0.275, 0.069, respectively). Cardiac b-type natriuretic peptide levels were increased in all hypoxia groups compared with normal (P < 0.001), but significantly decreased from sham and untreated in both TRASCET groups (P < 0.0001-0.005). Heart tumor necrosis factor-alpha levels were significantly elevated in sham and TRASCET groups (P = 0.009, 0.002), but normalized in the untreated and TRASCET-primed groups (P = 0.256, 0.456). Lung transforming growth factor-beta levels were significantly increased in both sham and untreated groups (P < 0.001, 0.003), but normalized in both TRASCET groups (P = 0.567, 0.303). Similarly, lung endothelin-1 levels were elevated in sham and untreated groups (P < 0.001 for both), but normalized in both TRASCET groups (P = 0.367, 0.928). We conclude that TRASCET with MSCs decreases markers of fetal cardiac strain, insufficiency, and inflammation, as well as of pulmonary fibrosis and hypertension in the rodent model of IUGR.
Collapse
Affiliation(s)
- Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Gellisch M, Bablok M, Divvela SSK, Morosan-Puopolo G, Brand-Saberi B. Systemic Prenatal Stress Exposure through Corticosterone Application Adversely Affects Avian Embryonic Skin Development. BIOLOGY 2023; 12:biology12050656. [PMID: 37237470 DOI: 10.3390/biology12050656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023]
Abstract
Prenatal stress exposure is considered a risk factor for developmental deficits and postnatal behavioral disorders. While the effect of glucocorticoid-associated prenatal stress exposure has been comprehensively studied in many organ systems, there is a lack of in-depth embryological investigations regarding the effects of stress on the integumentary system. To approach this, we employed the avian embryo as a model organism and investigated the effects of systemic pathologically-elevated glucocorticoid exposure on the development of the integumentary system. After standardized corticosterone injections on embryonic day 6, we compared the stress-exposed embryos with a control cohort, using histological and immunohistochemical analyses as well as in situ hybridization. The overarching developmental deficits observed in the stress-exposed embryos were reflected through downregulation of both vimentin as well as fibronectin. In addition, a deficient composition in the different skin layers became apparent, which could be linked to a reduced expression of Dermo-1 along with significantly reduced proliferation rates. An impairment of skin appendage formation could be demonstrated by diminished expression of Sonic hedgehog. These results contribute to a more profound understanding of prenatal stress causing severe deficits in the integumentary system of developing organisms.
Collapse
Affiliation(s)
- Morris Gellisch
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Martin Bablok
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Satya Srirama Karthik Divvela
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
11
|
Hoodbhoy Z, Mohammed N, Nathani KR, Sattar S, Chowdhury D, Maskatia S, Tierney S, Hasan B, Das JK. The Impact of Maternal Preeclampsia and Hyperglycemia on the Cardiovascular Health of the Offspring: A Systematic Review and Meta-analysis. Am J Perinatol 2023; 40:363-374. [PMID: 33940650 DOI: 10.1055/s-0041-1728823] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The objective of this review was to assess the impact of maternal preeclampsia or hyperglycemia on the body composition and cardiovascular health in the offspring. STUDY DESIGN We conducted a systematic review utilizing PubMed, EBSCO, CINAHLPlus, Cochrane Library, and Web of Science to include all studies assessing the impact of preeclampsia/eclampsia and/or gestational/pregestational diabetes mellitus on the health of the offspring (children <10 years of age). The health measures included anthropometry, cardiac dimensions and function, and vascular function. We performed a meta-analysis using Review Manager software and computed net risk ratio (RR) with 95% confidence interval (CI) for dichotomous data and mean difference (MD) with 95% CI for continuous data. RESULTS There were 6,376 studies in total, of which 45 were included in the review and 40 in the meta-analysis. The results demonstrated higher birth weight (MD: 0.12 kg; 95% CI: 0.06-0.18) and systolic and diastolic blood pressure (BP; MD: 5.98 mm Hg; 95% CI: 5.64-6.32 and MD: 3.27 mm Hg; 95% CI: 0.65-5.89, respectively) in the offspring of mothers with gestational diabetes compared to controls. In contrast, the offspring of mothers with preeclampsia had lower birth weight (MD: -0.41 kg; 95% CI: -0.7 to -0.11); however, they had increased systolic (MD: 2.2 mm Hg; 95% CI: 1.28-3.12) and diastolic BP (MD: 1.41 mm Hg; 95% CI: 0.3-2.52) compared to controls. There is lack of data to conduct a meta-analysis of cardiac morphology, functional, and vascular imaging parameters. CONCLUSION These findings suggest that the in-utero milieu can have a permanent impact on the body composition and vascular health of the offspring. Future work warrants multicenter prospective studies to understand the mechanism and the actual effect of exposure to maternal hyperglycemia and high BP on the cardiovascular health of the offspring and long-term outcomes. KEY POINTS · Adverse in-utero exposures may have an impact on cardiovascular risk in children.. · Maternal hyperglycemia/preeclampsia lead to changes in birthweight and BP.. · Limited echocardiographic and vascular imaging data in these cohorts necessitates future work..
Collapse
Affiliation(s)
- Zahra Hoodbhoy
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Nuruddin Mohammed
- Department of Obstetrics and Gynecology, The Aga Khan University, Karachi, Pakistan
| | | | - Saima Sattar
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | | | - Shiraz Maskatia
- Department of Paediatrics, Stanford University, Stanford, California
| | - Seda Tierney
- Department of Paediatrics, Stanford University, Stanford, California
| | - Babar Hasan
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Jai K Das
- Division of Women and Child Health, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
12
|
Ramamoorthi Elangovan V, Saadat N, Ghnenis A, Padmanabhan V, Vyas AK. Developmental programming: adverse sexually dimorphic transcriptional programming of gestational testosterone excess in cardiac left ventricle of fetal sheep. Sci Rep 2023; 13:2682. [PMID: 36792653 PMCID: PMC9932081 DOI: 10.1038/s41598-023-29212-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Adverse in-utero insults during fetal life alters offspring's developmental trajectory, including that of the cardiovascular system. Gestational hyperandrogenism is once such adverse in-utero insult. Gestational testosterone (T)-treatment, an environment of gestational hyperandrogenism, manifests as hypertension and pathological left ventricular (LV) remodeling in adult ovine offspring. Furthermore, sexual dimorphism is noted in cardiomyocyte number and morphology in fetal life and at birth. This study investigated transcriptional changes and potential biomarkers of prenatal T excess-induced adverse cardiac programming. Genome-wide coding and non-coding (nc) RNA expression were compared between prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30 to 90 of gestation; Term: 147 days) and control ovine LV at day 90 fetus in both sexes. Prenatal T induced differential expression of mRNAs in the LV of female (2 down, 5 up) and male (3 down, 1 up) (FDR < 0.05, absolute log2 fold change > 0.5); pathways analysis demonstrated 205 pathways unique to the female, 382 unique to the male and 23 common pathways. In the male, analysis of ncRNA showed differential regulation of 15 lncRNAs (14 down, 1 up) and 27 snoRNAs (26 down and 1 up). These findings suggest sexual dimorphic modulation of cardiac coding and ncRNA with gestational T excess.
Collapse
Affiliation(s)
| | - Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Arpita K Vyas
- College of Medicine, California Northstate University, Elk Grove, CA, USA.
- Department of Pediatrics, Division of Pediatric Endocrinology, School of Medicine, Washington University, St Louis, MO, USA.
| |
Collapse
|
13
|
Ramli I, Posadino AM, Giordo R, Fenu G, Fardoun M, Iratni R, Eid AH, Zayed H, Pintus G. Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations. Antioxidants (Basel) 2023; 12:antiox12020341. [PMID: 36829900 PMCID: PMC9952837 DOI: 10.3390/antiox12020341] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Adverse pregnancy outcomes are considered significant health risks for pregnant women and their offspring during pregnancy and throughout their lifespan. These outcomes lead to a perturbated in-utero environment that impacts critical phases of the fetus's life and correlates to an increased risk of chronic pathological conditions, such as diabetes, obesity, and cardiovascular diseases, in both the mother's and adult offspring's life. The dietary intake of naturally occurring antioxidants promotes health benefits and disease prevention. In this regard, maternal dietary intake of polyphenolic antioxidants is linked to a reduced risk of maternal obesity and cardio-metabolic disorders, positively affecting both the fetus and offspring. In this work, we will gather and critically appraise the current literature highlighting the effect/s of the naturally occurring polyphenol antioxidant resveratrol on oxidative stress, inflammation, and other molecular and physiological phenomena associated with pregnancy and pregnancy conditions, such as gestational diabetes, preeclampsia, and preterm labor. The resveratrol impact on prenatal complications and pregnancy-associated structures, such as the fetus and placenta, will also be discussed. Finally, we will draw conclusions from the current knowledge and provide future perspectives on potentially exploiting resveratrol as a therapeutic tool in pregnancy-associated conditions.
Collapse
Affiliation(s)
- Iman Ramli
- Departement de Biologie Animale, Université des Frères Mentouri Constantine 1, Constantine 25000, Algeria
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Grazia Fenu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Manal Fardoun
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11-0236, Lebanon
| | - Rabah Iratni
- Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
14
|
Zhai X, Liu J, Yu M, Zhang Q, Li M, Zhao N, Liu J, Song Y, Ma L, Li R, Qiao Z, Zhao G, Wang R, Xiao X. Nontargeted metabolomics reveals the potential mechanism underlying the association between birthweight and metabolic disturbances. BMC Pregnancy Childbirth 2023; 23:14. [PMID: 36624413 PMCID: PMC9830726 DOI: 10.1186/s12884-023-05346-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
AIMS The aim of this study was to characterize the metabolites associated with small- and large-gestational-age newborns in maternal and cord blood, and to investigate potential mechanisms underlying the association between birthweight and metabolic disturbances. RESEARCH DESIGN AND METHODS We recorded detailed anthropometric data of mother-offspring dyads. Untargeted metabolomic assays were performed on 67 pairs of cord blood and maternal fasting plasma samples including 16 pairs of small-for-gestational (SGA, < 10th percentile) dyads, 28 pairs of appropriate-for-gestational (AGA, approximate 50 percentile) dyads, and 23 pairs of large-for-gestational (LGA, > 90th percentile) dyads. The association of metabolites with newborn birthweight was conducted to screen for metabolites with U-shaped and line-shaped distributions. The association of metabolites with maternal and fetal phenotypes was also performed. RESULTS We found 2 types of metabolites that changed in different patterns according to newborn birthweight. One type of metabolite exhibited a "U-shaped" trend of abundance fluctuation in the SGA-AGA-LGA groups. The results demonstrated that cuminaldehyde level was lower in the SGA and LGA groups, and its abundance in cord blood was negatively correlated with maternal BMI (r = -0.352 p = 0.009) and weight gain (r = -0.267 p = 0.043). 2-Methoxy-estradiol-17b 3-glucuronide, which showed enrichment in the SGA and LGA groups, was positively correlated with homocysteine (r = 0.44, p < 0.001) and free fatty acid (r = 0.42, p < 0.001) in maternal blood. Serotonin and 13(S)-HODE were the second type of metabolites, denoted as "line-shaped", which both showed increasing trends in the SGA-AGA-LGA groups in both maternal and cord blood and were both significantly positively correlated with maternal BMI before pregnancy. Moreover, cuminaldehyde, serotonin, 13(S)-HODE and some lipid metabolites showed a strong correlation between maternal and cord blood. CONCLUSIONS These investigations demonstrate broad-scale metabolomic differences associated with newborn birthweight in both pregnant women and their newborns. The U-shaped metabolites associated with both the SGA and LGA groups might explain the U-shaped association between birthweight and metabolic dysregulation. The line-shaped metabolites might participate in intrauterine growth regulation. These observations might help to provide new insights into the insulin resistance and the risk of metabolic disturbance of SGA and LGA babies in adulthood and might identify potential new markers for adverse newborn outcomes in pregnant women.
Collapse
Affiliation(s)
- Xiao Zhai
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Jieying Liu
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China ,grid.413106.10000 0000 9889 6335Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Miao Yu
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Qian Zhang
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Ming Li
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Nan Zhao
- grid.413106.10000 0000 9889 6335Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Juntao Liu
- grid.413106.10000 0000 9889 6335Department of Obstetrics & Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Yingna Song
- grid.413106.10000 0000 9889 6335Department of Obstetrics & Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Liangkun Ma
- grid.413106.10000 0000 9889 6335Department of Obstetrics & Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Rongrong Li
- grid.413106.10000 0000 9889 6335Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Zongxu Qiao
- grid.478131.80000 0004 9334 6499Department of Obstetrics & Gynecology, Xingtai People’s Hospital, Xingtai, Hebei 054000 People’s Republic of China
| | - Guifen Zhao
- grid.478131.80000 0004 9334 6499Department of Obstetrics & Gynecology, Xingtai People’s Hospital, Xingtai, Hebei 054000 People’s Republic of China
| | - Ruiping Wang
- grid.478131.80000 0004 9334 6499Department of Obstetrics & Gynecology, Xingtai People’s Hospital, Xingtai, Hebei 054000 People’s Republic of China
| | - Xinhua Xiao
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
15
|
Lucas-Herald AK, Touyz RM. Androgens and Androgen Receptors as Determinants of Vascular Sex Differences Across the Lifespan. Can J Cardiol 2022; 38:1854-1864. [PMID: 36156286 DOI: 10.1016/j.cjca.2022.09.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022] Open
Abstract
Androgens, including testosterone and its more potent metabolite dihydrotestosterone, exert multiple actions in the body. Physiologically, they play a critical role in male sex development. In addition, they influence vascular function, including arterial vasodilation and mediation of myogenic tone. Androgens are produced from 9 weeks' gestation in the human fetal testis, as well as in small amounts by the adrenal glands. Serum concentrations vary according to age and sex. The vasculature is a target for direct actions of androgens, which bind to various sex hormone receptors expressed in endothelial and vascular smooth muscle cells. Androgens exert both vasoprotective and vasoinjurious effects, depending on multiple factors including sex-specific effects of androgens, heterogeneity of the vascular endothelium, differential expression of androgen and sex hormone receptors in endothelial and vascular smooth muscle cells, and the chronicity of androgen administration. Long-term administration of androgens induces vasoconstriction and influences endothelial permeability, whereas acute administration may have opposite effects. At the cellular level, androgens stimulate endothelial cell production of nitric oxide and inhibit proinflammatory signalling pathways, inducing vasorelaxation and vasoprotection. However, androgens also activate endothelial production of vasoconstrictors and stimulate recruitment of endothelial progenitor cells. In humans, both androgen deficiency and androgen excess are associated with increased cardiovascular morbidity and mortality. This review discusses how androgens modulate vascular sex differences across the lifespan by considering the actions and production of androgens in both sexes and describes how cardiovascular risk is altered as levels of androgens change with aging.
Collapse
Affiliation(s)
- Angela K Lucas-Herald
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom.
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montréal, Québec, Canada.
| |
Collapse
|
16
|
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
|
17
|
Steller JG, Gumina D, Driver C, Peek E, Galan HL, Reeves S, Hobbins JC. Patterns of Brain Sparing in a Fetal Growth Restriction Cohort. J Clin Med 2022; 11:jcm11154480. [PMID: 35956097 PMCID: PMC9369342 DOI: 10.3390/jcm11154480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022] Open
Abstract
Objective: Our objective was to compare differences in Doppler blood flow in four fetal intracranial blood vessels in fetuses with late-onset fetal growth restriction (FGR) vs. those with small for gestational age (SGA). Methods: Fetuses with estimated fetal weight (EFW) <10th percentile were divided into SGA (n = 30) and FGR (n = 51) via Delphi criteria and had Doppler waveforms obtained from the middle cerebral artery (MCA), anterior cerebral artery (ACA), posterior cerebral artery (PCA), and vertebral artery (VA). A pulsatility index (PI) <5th centile was considered “abnormal”. Outcomes included birth metrics and neonatal intensive care unit (NICU) admission. Results: There were more abnormal cerebral vessel PIs in the FGR group versus the SGA group (36 vs. 4; p = 0.055). In FGR, ACA + MCA vessel abnormalities outnumbered PCA + VA abnormalities. All 8 fetuses with abnormal VA PIs had at least one other abnormal vessel. Fetuses with abnormal VA PIs had lower BW (1712 vs. 2500 g; p < 0.0001), delivered earlier (35.22 vs. 37.89 wks; p = 0.0052), and had more admissions to the NICU (71.43% vs. 24.44%; p = 0.023). Conclusions: There were more anterior vessels showing vasodilation than posterior vessels, but when the VA was abnormal, the fetuses were more severely affected clinically than those showing normal VA PIs.
Collapse
Affiliation(s)
- Jon G. Steller
- Correspondence: ; Tel.: +1-714-456-6810 or +1-559-360-8545
| | | | | | | | | | | | | |
Collapse
|
18
|
Steller JG, Gumina D, Driver C, Palmer C, Brown LD, Reeves S, Hobbins JC, Galan HL. 3D Fractional Limb Volume Identifies Reduced Subcutaneous and Lean Mass in Fetal Growth Restriction. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:1623-1632. [PMID: 34580892 DOI: 10.1002/jum.15841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/30/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES Fetal 2D and 3D fractional limb volume (FLV) measurements by ultrasound can detect fetal lean and subcutaneous mass and possibly percent body fat. Our objectives were to 1) compare FLV measurements in fetuses with fetal growth restriction (FGR) versus small for gestational age (SGA) defined by the International Society of Ultrasound in Obstetrics and Gynecology (ISUOG)-supported international Delphi consensus and 2) correlate FLV findings with birth metrics. We hypothesize that FLV measurements will be significantly smaller in FGR versus SGA fetuses and will correlate closer with Ponderal index (PIx) in the neonate than abdominal circumference (AC). METHODS Patients were categorized as FGR or SGA as defined by ISUOG. Total thigh volume (TTV), volumes of lean mass (LMV), and fat mass volume (FMV) were calculated from 3D acquisitions. Measurements were compared between groups and correlated with birthweight (BW) and PIx (BW/crown-heal length). RESULTS The FGR group (n = 37) delivered earlier (37/2 versus 38/0; P = .0847), were lighter (2.2 kg versus 2.6 kg; P = .0003) and had lower PIx (0.023 versus 0.025; P = .0013) than SGAs (n = 22). FGRs had reduced TTV (40.6 versus 48.4 cm3 ; P = .0164), FMV (20.8 versus 25.3 cm3 ; P = .0413), and LMV (19.8 versus 23.1 cm3 ; P = .0387). AC had the highest area under the curve (0.69) for FGR. FMV was more strongly associated with PIx than the AC (P = .0032). CONCLUSIONS The AC and FLV measurements were significantly reduced in FGR fetuses compared to SGAs. While the AC outperformed FLV in predicting FGR, the FLV correlated best with PIx, which holds investigative promise.
Collapse
Affiliation(s)
- Jonathan G Steller
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of California, Irvine, Orange, CA, USA
| | - Diane Gumina
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Camille Driver
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Claire Palmer
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Laura D Brown
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Shane Reeves
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - John C Hobbins
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Henry L Galan
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
19
|
Heath-Freudenthal A, Toledo-Jaldin L, von Alvensleben I, Lazo-Vega L, Mizutani R, Stalker M, Yasini H, Mendizabal F, Madera JD, Mundo W, Castro-Monrroy M, Houck JA, Moreno-Aramayo A, Miranda-Garrido V, Su EJ, Giussani DA, Abman SH, Moore LG, Julian CG. Vascular Disorders of Pregnancy Increase Susceptibility to Neonatal Pulmonary Hypertension in High-Altitude Populations. Hypertension 2022; 79:1286-1296. [PMID: 35437031 PMCID: PMC9098686 DOI: 10.1161/hypertensionaha.122.19078] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preeclampsia and fetal growth restriction increase cardiopulmonary disease risk for affected offspring and occur more frequently at high-altitude (≥2500 m). Retrospective studies indicate that birth to a preeclampsia woman at high altitude increases the risk of pulmonary hypertension (PH) in later life. This prospective study asked whether preeclampsia with or without fetal growth restriction exaggerated fetal hypoxia and impaired angiogenesis in the fetal lung, leading to neonatal cardiopulmonary circulation abnormalities and neonatal or infantile PH. METHODS AND RESULTS We studied 79 maternal-infant pairs (39 preeclampsia, 40 controls) in Bolivia (3600-4100 m). Cord blood erythropoietin, hemoglobin, and umbilical artery and venous blood gases were measured as indices of fetal hypoxia. Maternal and cord plasma levels of angiogenic (VEGF [vascular endothelial growth factor]) and antiangiogenic (sFlt1 [soluble fms-like tyrosine kinase]) factors were determined. Postnatal echocardiography (1 week and 6-9 months) assessed pulmonary hemodynamics and PH. Preeclampsia augmented fetal hypoxia and increased the risk of PH in the neonate but not later in infancy. Pulmonary abnormalities were confined to preeclampsia cases with fetal growth restriction. Maternal and fetal plasma sFlt1 levels were higher in preeclampsia than controls and positively associated with PH. CONCLUSIONS The effect of preeclampsia with fetal growth restriction to increase fetal hypoxia and sFlt1 levels may impede normal development of the pulmonary circulation at high altitude, leading to adverse neonatal pulmonary vascular outcomes. Our observations highlight important temporal windows for the prevention of pulmonary vascular disease among babies born to highland residents or those with exaggerated hypoxia in utero or newborn life.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hussna Yasini
- College of Liberal Arts and Sciences, University of Colorado Denver, Denver, Colorado
| | | | - Jesus Dorado Madera
- College of Liberal Arts and Sciences, University of Colorado Denver, Denver, Colorado
| | - William Mundo
- University of Colorado School of Medicine, Aurora, Colorado
| | | | - Julie A. Houck
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | | | | | - Emily J. Su
- Departments of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Dino A. Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Steven H. Abman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Lorna G. Moore
- Departments of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Colleen G. Julian
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
20
|
Ji B, Lei J, Xu T, Zhao M, Cai H, Qiu J, Gao Q. Effects of prenatal hypoxia on placental glucocorticoid barrier: mechanistic insight from experiments in rats. Reprod Toxicol 2022; 110:78-84. [DOI: 10.1016/j.reprotox.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022]
|
21
|
The role of plasminogen activator inhibitor-1 in gynecological and obstetrical diseases: an update review. J Reprod Immunol 2022; 150:103490. [DOI: 10.1016/j.jri.2022.103490] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/21/2022]
|
22
|
Guo S, Wu X, Pei J, Wang X, Bao P, Xiong L, Chu M, Liang C, Yan P, Guo X. Genome-wide CNV analysis reveals variants associated with high-altitude adaptation and meat traits in Qaidam cattle. ELECTRON J BIOTECHN 2021. [DOI: 10.1016/j.ejbt.2021.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
23
|
Epigenetic processes during preeclampsia and effects on fetal development and chronic health. Clin Sci (Lond) 2021; 135:2307-2327. [PMID: 34643675 DOI: 10.1042/cs20190070] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/08/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
Preeclampsia (PE), the leading cause of maternal and fetal morbidity and mortality, is associated with poor fetal growth, intrauterine growth restriction (IUGR) and low birth weight (LBW). Offspring of women who had PE are at increased risk for cardiovascular (CV) disease later in life. However, the exact etiology of PE is unknown. Moreover, there are no effective interventions to treat PE or alleviate IUGR and the developmental origins of chronic disease in the offspring. The placenta is critical to fetal growth and development. Epigenetic regulatory processes such as histone modifications, microRNAs and DNA methylation play an important role in placental development including contributions to the regulation of trophoblast invasion and remodeling of the spiral arteries. Epigenetic processes that lead to changes in placental gene expression in PE mediate downstream effects that contribute to the development of placenta dysfunction, a critical mediator in the onset of PE, impaired fetal growth and IUGR. Therefore, this review will focus on epigenetic processes that contribute to the pathogenesis of PE and IUGR. Understanding the epigenetic mechanisms that contribute to normal placental development and the initiating events in PE may lead to novel therapeutic targets in PE that improve fetal growth and mitigate increased CV risk in the offspring.
Collapse
|
24
|
Lee K, Hardy DB. Metabolic Consequences of Gestational Cannabinoid Exposure. Int J Mol Sci 2021; 22:9528. [PMID: 34502436 PMCID: PMC8430813 DOI: 10.3390/ijms22179528] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
Up to 20% of pregnant women ages 18-24 consume cannabis during pregnancy. Moreover, clinical studies indicate that cannabis consumption during pregnancy leads to fetal growth restriction (FGR), which is associated with an increased risk of obesity, type II diabetes (T2D), and cardiovascular disease in the offspring. This is of great concern considering that the concentration of Δ9- tetrahydrocannabinol (Δ9-THC), a major psychoactive component of cannabis, has doubled over the last decade and can readily cross the placenta and enter fetal circulation, with the potential to negatively impact fetal development via the endocannabinoid (eCB) system. Cannabis exposure in utero could also lead to FGR via placental insufficiency. In this review, we aim to examine current pre-clinical and clinical findings on the direct effects of exposure to cannabis and its constituents on fetal development as well as indirect effects, namely placental insufficiency, on postnatal metabolic diseases.
Collapse
Affiliation(s)
- Kendrick Lee
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street, London, ON N6A 5C1, Canada;
- The Children’s Health Research Institute, The Lawson Health Research Institute, London, ON N6A 5C1, Canada
| | - Daniel B. Hardy
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street, London, ON N6A 5C1, Canada;
- The Children’s Health Research Institute, The Lawson Health Research Institute, London, ON N6A 5C1, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada
| |
Collapse
|
25
|
Fourman LT, Mueller SB, Boutin A, Zheng I, Pan CS, Gerard ME, Stanley TL, Roberts DJ. Placental Vascular Abnormalities in Association With Prenatal and Long-Term Health Characteristics Among HIV-Exposed Uninfected Adolescents and Young Adults. J Acquir Immune Defic Syndr 2021; 88:103-109. [PMID: 34034303 PMCID: PMC8373807 DOI: 10.1097/qai.0000000000002734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV-exposed uninfected (HEU) individuals are predisposed to adverse health outcomes, which in part may stem from the influence of an altered intrauterine milieu on fetal programming. The placenta serves as a readout for the effects of the maternal environment on the developing fetus and may itself contribute to the pathogenesis of disease. SETTING US academic health system. METHODS We leveraged a previously established registry-based cohort of HEU adolescents and young adults to identify 26 subjects for whom placental histopathology was available. We further obtained placental tissue from 29 HIV-unexposed pregnancies for comparison. We examined differences in placental histopathology between the groups and related villous vascularity in the HEU group to prenatal maternal characteristics and long-term health outcomes. RESULTS Placentas from HEU pregnancies demonstrated a higher blood vessel count per villus as compared with controls (5.9 ± 1.0 vs. 5.4 ± 0.8; P = 0.05), which was independent of maternal prenatal age, race, body mass index, smoking status, hemoglobin, and gestational age. Furthermore, within the HEU group, lower CD4+ T-cell count during pregnancy was associated with greater placental vascularity (r = -0.44; P = 0.03). No significant relationships were observed between placental blood vessel count per villus and body mass index z-score or reactive airway disease among HEU individuals later in life. CONCLUSIONS Placentas from HEU pregnancies demonstrated increased villous vascularity compared with HIV-unexposed controls in proportion to the severity of maternal immune dysfunction. Further studies are needed to examine intrauterine exposure to hypoxia as a potential mechanism of fetal programming in HIV.
Collapse
Affiliation(s)
- Lindsay T. Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sarah B. Mueller
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Autumn Boutin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Isabel Zheng
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Chelsea S. Pan
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Marisa E. Gerard
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Takara L. Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Pediatric Endocrinology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Drucilla J. Roberts
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Looi K, Kicic A, Noble PB, Wang KCW. Intrauterine growth restriction predisposes to airway inflammation without disruption of epithelial integrity in postnatal male mice. J Dev Orig Health Dis 2021; 12:496-504. [PMID: 32799948 DOI: 10.1017/s2040174420000744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Evidence from animal models demonstrate that intrauterine growth restriction (IUGR) alters airway structure and function which may affect susceptibility to disease. Airway inflammation and dysregulated epithelial barrier properties are features of asthma which have not been examined in the context of IUGR. This study used a maternal hypoxia-induced IUGR mouse model to assess lung-specific and systemic inflammation and airway epithelial tight junctions (TJs) protein expression. Pregnant BALB/c mice were housed under hypoxic conditions (10.5% O2) from gestational day (GD) 11 to 17.5 (IUGR group; term, GD 21). Following hypoxic exposure, mice were returned to a normoxic environment (21% O2). A Control group was housed under normoxic conditions throughout pregnancy. Offspring weights were recorded at 2 and 8 weeks of age and euthanized for bronchoalveolar lavage (BAL) and peritoneal cavity fluid collection for inflammatory cells counts. From a separate group of mice, right lungs were collected for Western blotting of TJs proteins. IUGR offspring had greater inflammatory cells in the BAL fluid but not in peritoneal fluid compared with Controls. At 8 weeks of age, interleukin (IL)-2, IL-13, and eotaxin concentrations were higher in male IUGR compared with male Control offspring but not in females. IUGR had no effect on TJs protein expression. Maternal hypoxia-induced IUGR increases inflammatory cells in the BAL fluid of IUGR offspring with no difference in TJs protein expression. Increased cytokine release, specific to the lungs of IUGR male offspring, indicates that both IUGR and sex can influence susceptibility to airway disease.
Collapse
Affiliation(s)
- Kevin Looi
- Telethon Kids Institute, The University of Western Australia, Crawley, WA6009, Australia
- School of Public Health, Curtin University, Bentley, WA6102, Australia
| | - Anthony Kicic
- Telethon Kids Institute, The University of Western Australia, Crawley, WA6009, Australia
- School of Public Health, Curtin University, Bentley, WA6102, Australia
- Faculty of Health and Medical Science, The University of Western Australia, Crawley, WA6009, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Crawley, WA6009, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, WA6009, Australia
| | - Kimberley C W Wang
- Telethon Kids Institute, The University of Western Australia, Crawley, WA6009, Australia
- School of Human Sciences, The University of Western Australia, Crawley, WA6009, Australia
| |
Collapse
|
27
|
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: 4.7] [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
|
28
|
Mundo W, Wolfson G, Moore LG, Houck JA, Park D, Julian CG. Hypoxia-induced inhibition of mTORC1 activity in the developing lung: a possible mechanism for the developmental programming of pulmonary hypertension. Am J Physiol Heart Circ Physiol 2021; 320:H980-H990. [PMID: 33416457 PMCID: PMC7988757 DOI: 10.1152/ajpheart.00520.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Perinatal hypoxia induces permanent structural and functional changes in the lung and its pulmonary circulation that are associated with the development of pulmonary hypertension (PH) in later life. The mechanistic target of the rapamycin (mTOR) pathway is vital for fetal lung development and is implicated in hypoxia-associated PH, yet its involvement in the developmental programming of PH remains unclear. Pregnant C57/BL6 dams were placed in hyperbaric (760 mmHg) or hypobaric chambers during gestation (505 mmHg, day 15 through postnatal day 4) or from weaning through adulthood (420 mmHg, postnatal day 21 through 8 wk). Pulmonary hemodynamics and right ventricular systolic pressure (RVSP) were measured at 8 wk. mTOR pathway proteins were assessed in fetal (day 18.5) and adult lung (8 wk). Perinatal hypoxia induced PH during adulthood, even in the absence of a sustained secondary hypoxic exposure, as indicated by reduced pulmonary artery acceleration time (PAAT) and peak flow velocity through the pulmonary valve, as well as greater RVSP, right ventricular (RV) wall thickness, and RV/left ventricular (LV) weight. Such effects were independent of increased blood viscosity. In fetal lung homogenates, hypoxia reduced the expression of critical downstream mTOR targets, most prominently total and phosphorylated translation repressor protein (4EBP1), as well as vascular endothelial growth factor, a central regulator of angiogenesis in the fetal lung. In contrast, adult offspring of hypoxic dams tended to have elevated p4EBP1 compared with controls. Our data suggest that inhibition of mTORC1 activity in the fetal lung as a result of gestational hypoxia may interrupt pulmonary vascular development and thereby contribute to the developmental programming of PH.NEW & NOTEWORTHY We describe the first study to evaluate a role for the mTOR pathway in the developmental programming of pulmonary hypertension. Our findings suggest that gestational hypoxia impairs mTORC1 activation in the fetal lung and may impede pulmonary vascular development, setting the stage for pulmonary vascular disease in later life.
Collapse
Affiliation(s)
- William Mundo
- School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Gabriel Wolfson
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Do Park
- School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
29
|
Ganguly E, Kirschenman R, Spaans F, Holody CD, Phillips TEJ, Case CP, Cooke CLM, Murphy MP, Lemieux H, Davidge ST. Nanoparticle-encapsulated antioxidant improves placental mitochondrial function in a sexually dimorphic manner in a rat model of prenatal hypoxia. FASEB J 2021; 35:e21338. [PMID: 33428278 DOI: 10.1096/fj.202002193r] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022]
Abstract
Pregnancy complications associated with prenatal hypoxia lead to increased placental oxidative stress. Previous studies suggest that prenatal hypoxia can reduce mitochondrial respiratory capacity and mitochondrial fusion, which could lead to placental dysfunction and impaired fetal development. We developed a placenta-targeted treatment strategy using a mitochondrial antioxidant, MitoQ, encapsulated into nanoparticles (nMitoQ) to reduce placental oxidative stress and (indirectly) improve fetal outcomes. We hypothesized that, in a rat model of prenatal hypoxia, nMitoQ improves placental mitochondrial function and promotes mitochondrial fusion in both male and female placentae. Pregnant rats were treated with saline or nMitoQ on gestational day (GD) 15 and exposed to normoxia (21% O2 ) or hypoxia (11% O2 ) from GD15-21. On GD21, male and female placental labyrinth zones were collected for mitochondrial respirometry assessments, mitochondrial content, and markers of mitochondrial biogenesis, fusion and fission. Prenatal hypoxia reduced complex IV activity and fusion in male placentae, while nMitoQ improved complex IV activity in hypoxic male placentae. In female placentae, prenatal hypoxia decreased respiration through the S-pathway (complex II) and increased N-pathway (complex I) respiration, while nMitoQ increased fusion in hypoxic female placentae. No changes in mitochondrial content, biogenesis or fission were found. In conclusion, nMitoQ improved placental mitochondrial function in male and female placentae from fetuses exposed to prenatal hypoxia, which may contribute to improved placental function. However, the mechanisms (ie, changes in mitochondrial respiratory capacity and mitochondrial fusion) were distinct between the sexes. Treatment strategies targeted against placental oxidative stress could improve placental mitochondrial function in complicated pregnancies.
Collapse
Affiliation(s)
- Esha Ganguly
- Department of Physiology, University of Alberta, Edmonton, AB, Canada.,Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Claudia D Holody
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada.,Department of Paediatrics, University of Alberta, Edmonton, AB, Canada.,Faculty Saint-Jean, University of Alberta, Edmonton, AB, Canada
| | | | - C Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol, UK
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Keith Peters Building, University of Cambridge, Cambridge, UK
| | - Hélène Lemieux
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada.,Faculty Saint-Jean, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, AB, Canada.,Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Lee K, Laviolette SR, Hardy DB. Exposure to Δ9-tetrahydrocannabinol during rat pregnancy leads to impaired cardiac dysfunction in postnatal life. Pediatr Res 2021; 90:532-539. [PMID: 33879850 PMCID: PMC8519775 DOI: 10.1038/s41390-021-01511-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Cannabis use in pregnancy leads to fetal growth restriction (FGR), but the long-term effects on cardiac function in the offspring are unknown, despite the fact that fetal growth deficits are associated with an increased risk of developing postnatal cardiovascular disease. We hypothesize that maternal exposure to Δ9-tetrahydrocannabinol (Δ9-THC) during pregnancy will impair fetal development, leading to cardiac dysfunction in the offspring. METHODS Pregnant Wistar rats were randomly selected and administered 3 mg/kg of Δ9-THC or saline as a vehicle daily via intraperitoneal injection from gestational days 6 to 22, followed by echocardiogram analysis of cardiac function on offspring at postnatal days 1 and 21. Heart tissue was harvested from the offspring at 3 weeks for molecular analysis of cardiac remodelling. RESULTS Exposure to Δ9-THC during pregnancy led to FGR with a significant decrease in heart-to-body weight ratios at birth. By 3 weeks, pups exhibited catch-up growth associated with significantly greater left ventricle anterior wall thickness with a decrease in cardiac output. Moreover, these Δ9-THC-exposed offsprings exhibited increased expression of collagen I and III, decreased matrix metallopeptidase-2 expression, and increased inactivation of glycogen synthase kinase-3β, all associated with cardiac remodelling. CONCLUSIONS Collectively, these data suggest that Δ9-THC-exposed FGR offspring undergo postnatal catch-up growth concomitant with cardiac remodelling and impaired cardiac function early in life. IMPACT To date, the long-term effects of perinatal Δ9-THC (the main psychoactive component) exposure on the cardiac function in the offspring remain unknown. We demonstrated, for the first time, that exposure to Δ9-THC alone during rat pregnancy results in significantly smaller hearts relative to body weight. These Δ9-THC-exposed offsprings exhibited postnatal catch-up growth concomitant with cardiac remodelling and impaired cardiac function. Given the increased popularity of cannabis use in pregnancy along with rising Δ9-THC concentrations, this study, for the first time, identifies the risk of perinatal Δ9-THC exposure on early postnatal cardiovascular health.
Collapse
Affiliation(s)
- Kendrick Lee
- grid.39381.300000 0004 1936 8884Department of Physiology and Pharmacology, Western University, London, ON Canada
| | - Steven R. Laviolette
- grid.39381.300000 0004 1936 8884Department of Anatomy and Cell Biology, Western University, London, ON Canada
| | - Daniel B. Hardy
- grid.39381.300000 0004 1936 8884Department of Physiology and Pharmacology, Western University, London, ON Canada ,grid.39381.300000 0004 1936 8884Departments of Obstetrics and Gynecology, Children’s Health Research Institute, Lawson, Health Research Institute, Western University, London, ON Canada
| |
Collapse
|
31
|
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: 3.0] [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
|
32
|
Sorensen DW, Carreon D, Williams JM, Pearce WJ. Hypoxic modulation of fetal vascular MLCK abundance, localization, and function. Am J Physiol Regul Integr Comp Physiol 2021; 320:R1-R18. [PMID: 33112654 PMCID: PMC7847055 DOI: 10.1152/ajpregu.00212.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 12/26/2022]
Abstract
Changes in vascular contractility are among the most important physiological effects of acute and chronic fetal hypoxia. Given the essential role of myosin light-chain kinase (MLCK) in smooth muscle contractility and its heterogeneous distribution, this study explores the hypothesis that subcellular changes in MLCK distribution contribute to hypoxic modulation of fetal carotid artery contractility. Relative to common carotid arteries from normoxic term fetal lambs (FN), carotids from fetal lambs gestated at high altitude (3,802 m) (FH) exhibited depressed contractility without changes in MLCK mRNA or protein abundance. Patterns of confocal colocalization of MLCK with α-actin and 20-kDa regulatory myosin light chain (MLC20) enabled calculation of subcellular MLCK fractions: 1) colocalized with the contractile apparatus, 2) colocalized with α-actin distant from the contractile apparatus, and 3) not colocalized with α-actin. Chronic hypoxia did not affect MLCK abundance in the contractile fraction, despite a concurrent decrease in contractility. Organ culture for 72 h under 1% O2 decreased total MLCK abundance in FN and FH carotid arteries, but decreased the contractile MLCK abundance only in FH carotid arteries. Correspondingly, culture under 1% O2 depressed contractility more in FH than FN carotid arteries. In addition, hypoxia appeared to attenuate ubiquitin-independent proteasomal degradation of MLCK, as reported for other proteins. In aggregate, these results demonstrate that the combination of chronic hypoxia followed by hypoxic culture can induce MLCK translocation among at least three subcellular fractions with possible influences on contractility, indicating that changes in MLCK distribution are a significant component of fetal vascular responses to hypoxia.
Collapse
Affiliation(s)
- Dane W Sorensen
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Desirelys Carreon
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - James M Williams
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - William J Pearce
- Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
33
|
Do V, Eckersley L, Lin L, Davidge ST, Stickland MK, Ojala T, Serrano-Lomelin J, Hornberger LK. Persistent Aortic Stiffness and Left Ventricular Hypertrophy in Children of Diabetic Mothers. CJC Open 2020; 3:345-353. [PMID: 33778451 PMCID: PMC7985002 DOI: 10.1016/j.cjco.2020.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 01/11/2023] Open
Abstract
Background Fetuses of diabetic mothers develop left ventricular (LV) hypertrophy and are at increased long-term risk of cardiovascular disease. In our previous longitudinal study from midgestation to late infancy we showed persistence of LV hypertrophy and increased aortic stiffness compared with infants of healthy mothers, the latter of which correlated with third trimester maternal hemoglobin A1c. In the present study, we reexamined the same cohort in early childhood to determine if these cardiovascular abnormalities persisted. Methods Height, weight, and right arm blood pressure were recorded. A full functional and structural echocardiogram was performed with offline analysis of LV posterior wall and interventricular septal diastolic thickness (IVSd), systolic and diastolic function, and aortic pulse wave velocity. Vascular reactivity was assessed using digital thermal monitoring. Participants also completed a physical activity questionnaire. Results Twenty-five children of diabetic mothers (CDMs) and 20 children from healthy pregnancies (mean age, 5.6 ± 1.7 and 5.3 ± 1.3 years, respectively; P = not significant) were assessed. Compared with controls, IVSd z score was increased in CDMs (1.2 ± 0.6 vs 0.5 ± 0.3, respectively; P = 0.006), with one-fifth having a z score of more than +2.0. Aortic pulse wave velocity was increased in CDMs (3.2 ± 0.6 m/s vs 2.2 ± 0.4 m/s; P = 0.001), and correlated with IVSd z score (R2 = 0.81; P = 0.001) and third trimester maternal A1c (R2 = 0.65; P < 0.0001). Body surface area, height, weight, blood pressure, vascular reactivity, and physical activity scores did not differ between groups. Our longitudinal analysis showed that individuals with greater IVSd, and aortic stiffness in utero, early and late infancy also tended to have greater measures in early childhood (P < 0.001 and P < 0.0001, respectively). Conclusions CDMs show persistently increased interventricular septal thickness and aortic stiffness in early childhood.
Collapse
Affiliation(s)
- Victor Do
- Fetal and Neonatal Cardiology Program, Division of Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Luke Eckersley
- Fetal and Neonatal Cardiology Program, Division of Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Lily Lin
- Fetal and Neonatal Cardiology Program, Division of Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sandra T Davidge
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael K Stickland
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tiina Ojala
- University of Helsinki, Helsinki Children's Hospital, Helsinki, Finland
| | | | - Lisa K Hornberger
- Fetal and Neonatal Cardiology Program, Division of Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
de Almeida Silva LC, de Oliveira AC, Cavalcante-Silva V, Franco MDC, D'Almeida V. Hyperlipidic diet affects body composition and induces anxiety-like behaviour in intrauterine growth-restricted adult mice. Exp Physiol 2020; 105:2061-2072. [PMID: 33098335 DOI: 10.1113/ep088859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/16/2020] [Indexed: 12/31/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the effect in male and female offspring of a protein-deficient diet producing intrauterine growth restriction (IUGR) in maternal mice on morphometric, metabolic and behavioural parameters before and after a challenge with a fat diet? What is the main finding and its importance? Male and female mice presented different growth trajectories after birth. IUGR favoured increased adiposity in male mice, and high-fat diet-induced anxiety-like behaviour in female mice. ABSTRACT As there is sexual dimorphism in the response to maternal manipulations, we aimed to analyse the effects of intrauterine growth restriction (IUGR) in both sexes on morphometric, metabolic and behavioural parameters throughout postnatal development, and after challenge with a hyperlipidic diet. Female Swiss mice (n = 59) were distributed into two groups (SD: standard diet, n = 26; and PDD: isocaloric protein-deficient diet, n = 33), 2 weeks before mating and during the gestational period. After birth, offspring from SD and PDD dams were cross-fostered and nurtured by SD dams until postnatal day (PND) 28. At PND 60 all animals were challenged with a hypercaloric diet for 4 weeks. Offspring birth weight was significantly reduced in the PDD group compared to the SD group (P = 0.0001), but only male offspring presented a rapid catch-up during the first 21 days of development. Although no differences in body weight were observed between groups after the challenge with the hyperlipidic diet, an increase in the relative perigonadal white adipose tissue (P = 0.009) and a decrease in gross gastrocnemius muscle weight (P = 0.010) were observed in the PDD males. In relation to behavioural tests, there was an increase in locomotion in both sexes (P = 0.0001), and a decrease in female grooming (P = 0.006) in the PDD group. Additionally, females from the PDD group showed increased hyperlipidic food intake. In conclusion, IUGR affected both sexes, with females showing prominent behavioural modifications and males presenting altered body composition elicited by a hyperlipidic diet.
Collapse
Affiliation(s)
| | | | | | | | - Vânia D'Almeida
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
35
|
Woodman AG, Mah R, Keddie DL, Noble RMN, Holody CD, Panahi S, Gragasin FS, Lemieux H, Bourque SL. Perinatal iron deficiency and a high salt diet cause long-term kidney mitochondrial dysfunction and oxidative stress. Cardiovasc Res 2020; 116:183-192. [PMID: 30715197 DOI: 10.1093/cvr/cvz029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/08/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Perinatal iron deficiency (ID) alters developmental trajectories of offspring, predisposing them to cardiovascular dysfunction in later life. The mechanisms underlying this long-term programming of renal function have not been defined. We hypothesized perinatal ID causes hypertension and alters kidney metabolic function and morphology in a sex-dependent manner in adult offspring. Furthermore, we hypothesized these effects are exacerbated by chronic consumption of a high salt diet. METHODS AND RESULTS Pregnant Sprague Dawley rats were fed either an iron-restricted or replete diet prior to and throughout pregnancy. Adult offspring were fed normal or high salt diets for 6 weeks prior to experimentation at 6 months of age. Blood pressure (BP) was assessed via indwelling catheters in anaesthetized offspring; kidney mitochondrial function was assessed via high-resolution respirometry; reactive oxygen species and nitric oxide were quantified via fluorescence microscopy. Adult males, but not females, exhibited increased systolic BP due to ID (P = 0.01) and high salt intake (P = 0.02). In males, but not in females, medullary mitochondrial content was increased by high salt (P = 0.003), while succinate-dependent respiration was reduced by ID (P < 0.05). The combination of perinatal ID and high salt reduced complex IV activity in the cortex of males (P = 0.01). Perinatal ID increased cytosolic superoxide generation (P < 0.001) concomitant with reduced nitric oxide bioavailability (P < 0.001) in male offspring, while high salt increased mitochondrial superoxide in the medulla (P = 0.04) and cytosolic superoxide within the cortex (P = 0.01). Male offspring exhibited glomerular basement membrane thickening (P < 0.05), increased collagen deposition (P < 0.05), and glomerular hypertrophy (interaction, P = 0.02) due to both perinatal ID and high salt. Female offspring exhibited no alterations in mitochondrial function or morphology due to either high salt or ID. CONCLUSION Perinatal ID causes long-term sex-dependent alterations in renal metabolic function and morphology, potentially contributing to hypertension and increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Andrew G Woodman
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Richard Mah
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Danae L Keddie
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Ronan M N Noble
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Claudia D Holody
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Sareh Panahi
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Ferrante S Gragasin
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Helene Lemieux
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Medicine, University of Alberta, Edmonton, Canada.,Faculty Saint-Jean, University of Alberta, Edmonton, Canada
| | - Stephane L Bourque
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
36
|
Chuang TD, Ansari A, Yu C, Sakurai R, Harb A, Liu J, Khorram O, Rehan VK. Mechanism underlying increased cardiac extracellular matrix deposition in perinatal nicotine-exposed offspring. Am J Physiol Heart Circ Physiol 2020; 319:H651-H660. [PMID: 32795172 DOI: 10.1152/ajpheart.00021.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although increased predisposition to cardiac fibrosis and cardiac dysfunction has been demonstrated in the perinatally nicotine-exposed heart, the underlying mechanisms remain unclear. With the use of a well-established rat model and cultured primary neonatal rat cardiac fibroblasts, the effect of perinatal nicotine exposure on offspring heart extracellular matrix deposition and the likely underlying mechanisms were investigated. Perinatal nicotine exposure resulted in increased collagen type I (COL1A1) and III (COL3A1) deposition along with a decrease in miR-29 family and an increase in long noncoding RNA myocardial infarction-associated transcript (MIAT) levels in offspring heart. Nicotine treatment of isolated primary neonatal rat cardiac fibroblasts suggested that these effects were mediated via nicotinic acetylcholine receptors including α7 and the induced collagens accumulation was reversed by a gain-of function of miR-29 family. Knockdown of MIAT resulted in increased miR-29 family and decreased COL1A1 and COL3A1 levels, suggesting nicotine-mediated MIAT induction as the underlying mechanism for nicotine-induced collagen deposition. Luciferase reporter assay and RNA immunoprecipitation studies showed an intense physical interaction between MIAT, miR-29 family, and argonaute 2, corroborating the mechanistic link between perinatal nicotine exposure and increased extracellular matrix deposition. Overall, perinatal nicotine exposure resulted in lower miR-29 family levels in offspring heart, while it elevated cardiac MIAT and collagen type I and III levels. These findings provide mechanistic basis for cardiac dysfunction in perinatal nicotine-exposed offspring and offer multiple novel potential therapeutic targets.NEW & NOTEWORTHY Using an established rat model and cultured primary neonatal cardiac fibroblasts, we show that nicotine mediated MIAT induction as the underlying mechanism for the excessive cardiac collagen deposition. These observations provide mechanistic basis for the increased predisposition to cardiac dysfunction following perinatal cigarette/nicotine exposure and offer novel potential therapeutic targets.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of and Obstetrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Aamir Ansari
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Celia Yu
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Reiko Sakurai
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Amir Harb
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Jie Liu
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Omid Khorram
- Department of and Obstetrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| |
Collapse
|
37
|
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: 1.8] [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
|
38
|
Abbas G, Shah S, Hanif M, Shah A, Rehman AU, Tahir S, Nayab K, Asghar A. The frequency of pulmonary hypertension in newborn with intrauterine growth restriction. Sci Rep 2020; 10:8064. [PMID: 32415157 PMCID: PMC7229189 DOI: 10.1038/s41598-020-65065-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/22/2020] [Indexed: 11/17/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a clinical definition applied to neonates born with clinical features of malnutrition and in-utero growth retardation irrespective of their birth weight percentile. This study was aimed to determine the frequency of pulmonary hypertension (PH) in neonates with IUGR. In this descriptive cross-sectional study, we followed 96 neonates with IUGR (≤28 days) and 38 neonates without IUGR born in the department of the neonatal intensive care unit children hospital complex Multan, Pakistan. We analyzed certain factors such as gender, gestational age (GA) (weeks), birth weight (BW in kg), weight percentile (WP) for GA, meconium aspiration syndrome (MAS), birth asphyxia (BA) and respiratory distress syndrome (RDS) for pulmonary hypertension (PH) in IUGR and non-IUGR group. GA was measured by the Ballard scoring system. Echocardiography was performed for all patients by the pediatric cardiologist to measure pulmonary arterial (PA) pressure using Bernoulli’s equation. Out of total 96 IUGR neonates, 33.3% (n = 32) suffered from PH, of which 65.3% (n = 18) were male and 43.7% (n = 14) were female. The percentages of IUGR neonates with BA, MAS and RDS were 34.4%, 18.8% and 22.9% respectively. The data were analyzed using the SPSS-16 software to test the statistical significance of the results. A p-value less than 0.05 was considered significant. When the chi-square test was applied, it depicted that MAS was significantly associated with PH in IUGR neonates (p = 0.0001) compared to non-IUGR neonates. Our findings suggested an increased chance of PH in IUGR neonates and MAS may be a strong factor.
Collapse
Affiliation(s)
- Ghulam Abbas
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences Government College University Faisalabad, Faisalabad, Pakistan
| | - Shahid Shah
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences Government College University Faisalabad, Faisalabad, Pakistan.
| | - Muhammad Hanif
- Department of Pharmaceutics, Faculty of Pharmacy, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Abid Shah
- Children hospital and the Institute of Child Health Multan, Multan, Pakistan
| | - Anees Ur Rehman
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, University Sains Penang, Sains Penang, Malaysia
| | - Sana Tahir
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences Government College University Faisalabad, Faisalabad, Pakistan
| | - Komal Nayab
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences Government College University Faisalabad, Faisalabad, Pakistan
| | - Arzoo Asghar
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
39
|
Identification of key HIF-1α target genes that regulate adaptation to hypoxic conditions in Tibetan chicken embryos. Gene 2020; 729:144321. [DOI: 10.1016/j.gene.2019.144321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022]
|
40
|
Kumar P, Morton JS, Shah A, Do V, Sergi C, Serrano‐Lomelin J, Davidge ST, Beker D, Levasseur J, Hornberger LK. Intrauterine exposure to chronic hypoxia in the rat leads to progressive diastolic function and increased aortic stiffness from early postnatal developmental stages. Physiol Rep 2020; 8:e14327. [PMID: 31960611 PMCID: PMC6971413 DOI: 10.14814/phy2.14327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM We sought to explore whether fetal hypoxia exposure, an insult of placental insufficiency, is associated with left ventricular dysfunction and increased aortic stiffness at early postnatal ages. METHODS Pregnant Sprague Dawley rats were exposed to hypoxic conditions (11.5% FiO2 ) from embryonic day E15-21 or normoxic conditions (controls). After delivery, left ventricular function and aortic pulse wave velocity (measure of aortic stiffness) were assessed longitudinally by echocardiography from day 1 through week 8. A mixed ANOVA with repeated measures was performed to compare findings between groups across time. Myocardial hematoxylin and eosin and picro-sirius staining were performed to evaluate myocyte nuclear shape and collagen fiber characteristics, respectively. RESULTS Systolic function parameters transiently increased following hypoxia exposure primarily at week 2 (p < .008). In contrast, diastolic dysfunction progressed following fetal hypoxia exposure beginning weeks 1-2 with lower early inflow Doppler velocities, and less of an increase in early to late inflow velocity ratios and annular and septal E'/A' tissue velocities compared to controls (p < .008). As further evidence of altered diastolic function, isovolumetric relaxation time was significantly shorter relative to the cardiac cycle following hypoxia exposure from week 1 onward (p < .008). Aortic stiffness was greater following hypoxia from day 1 through week 8 (p < .008, except week 4). Hypoxia exposure was also associated with altered nuclear shape at week 2 and increased collagen fiber thickness at week 4. CONCLUSION Chronic fetal hypoxia is associated with progressive LV diastolic dysfunction, which corresponds with changes in nuclear shape and collagen fiber thickness, and increased aortic stiffness from early postnatal stages.
Collapse
Affiliation(s)
- Praveen Kumar
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Jude S. Morton
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Amin Shah
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Victor Do
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Consolato Sergi
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonABCanada
| | - Jesus Serrano‐Lomelin
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
| | - Sandra T. Davidge
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Donna Beker
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Jody Levasseur
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Lisa K. Hornberger
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
41
|
Yan L, Wang Y, Zhang Z, Xu S, Ullah R, Luo X, Xu X, Ma X, Chen Z, Zhang L, Lv Y, Du L. Postnatal delayed growth impacts cognition but rescues programmed impaired pulmonary vascular development in an IUGR rat model. Nutr Metab Cardiovasc Dis 2019; 29:1418-1428. [PMID: 31653519 DOI: 10.1016/j.numecd.2019.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/19/2019] [Accepted: 08/23/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Intrauterine growth restriction (IUGR) is a state of slower fetal growth usually followed by a catch-up growth. Postnatal catch-up growth in IUGR models increases the incidence of pulmonary arterial hypertension in adulthood. Here, we hypothesize that the adverse pulmonary vascular consequences of IUGR may be improved by slowing down postnatal growth velocity. Meanwhile, cognitive function was also studied. METHODS AND RESULTS We established an IUGR rat model by restricting maternal food throughout gestation. After birth, pups were fed a regular or restricted diet during lactation by changing litter size. Thus, there were three experimental groups according to the dam/offspring diet: C/C (gold standard), IUGR with catch-up growth (R/C) and IUGR with delayed growth (R/D). In adulthood (14 weeks of age), we assessed pulmonary vascular development by hemodynamic measurement and immunohistochemistry. Our results showed that adult R/C offspring developed an elevated mean pulmonary arterial pressure (mPAP) and pulmonary arteriolar remodeling accompanied with decreased eNOS mRNA and protein expressions compared to C/C or R/D offspring. This suggested that delayed postnatal growth improved pulmonary circulation compared to postnatal catch-up growth. Conversely, adult R/D offspring performed poorly in cognition. Behavior test and electrophysiology results exhibited a reduced synaptic plasticity. Furthermore, decreased mRNA expression levels of the memory-related gene zif268 and transcription factor recruitment factor p300 in the hippocampus region were also observed in R/D group. CONCLUSION These findings indicate that delayed postnatal growth results in cognitive impairment, but it reverses elevations in mPAP induced by postnatal catch-up growth following IUGR.
Collapse
Affiliation(s)
- LingLing Yan
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Wang
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - ZiMing Zhang
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - ShanShan Xu
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Rahim Ullah
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - XiaoFei Luo
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - XueFeng Xu
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - XiaoLu Ma
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Chen
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - LiYan Zhang
- Fujian University of Medicine, NICU, Fuzhou Children's Hospital of Fujian Province, Fuzhou, 350005, Fujian Province, China
| | - Ying Lv
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - LiZhong Du
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Chen YH, Li L, Chen W, Liu ZB, Ma L, Gao XX, He JL, Wang H, Zhao M, Yang YY, Xu DX. Pre-pregnancy underweight and obesity are positively associated with small-for-gestational-age infants in a Chinese population. Sci Rep 2019; 9:15544. [PMID: 31664141 PMCID: PMC6820714 DOI: 10.1038/s41598-019-52018-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/10/2019] [Indexed: 01/12/2023] Open
Abstract
The association between suboptimal pre-pregnancy body mass index (BMI) and small-for-gestational-age (SGA) infants is not well defined. We investigated the association between pre-pregnancy BMI and the risk of SGA infants in a Chinese population. We performed a cohort study among 12029 mothers with a pregnancy. This cohort consisted of pregnant women that were: normal-weight (62.02%), underweight (17.09%), overweight (17.77%) and obese (3.12%). Birth sizes were reduced in the underweight and obese groups compared with the normal-weight group. Linear regression analysis indicated that birth size was positively associated with BMI in both the underweight and normal-weight groups. Further analysis showed that 12.74% of neonates were SGA infants in the underweight group, higher than 7.43% of neonates reported in the normal-weight group (adjusted RR = 1.92; 95% CI: 1.61, 2.30). Unexpectedly, 17.60% of neonates were SGA infants in the obese group, much higher than the normal-weight group (adjusted RR = 2.17; 95% CI: 1.57, 3.00). Additionally, 18.40% of neonates were large-for-gestational-age (LGA) infants in the obese group, higher than 7.26% of neonates reported in the normal-weight group (adjusted RR = 3.00; 95% CI: 2.21, 4.06). These results suggest that pre-pregnancy underweight increases the risk of SGA infants, whereas obesity increases the risks of not only LGA infants, but also SGA infants.
Collapse
Affiliation(s)
- Yuan Hua Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,School of Public Health, Anhui Medical University, Hefei, 230032, China. .,Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.
| | - Li Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zhi Bing Liu
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Li Ma
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Xing Xing Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jia Liu He
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Mei Zhao
- School of Nursing, Anhui Medical University, Hefei, 230032, China
| | - Yuan Yuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - De Xiang Xu
- School of Public Health, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
43
|
Reckelhoff JF, LaMarca B, Garovic VD, Alexander BT. Human Umbilical Venous Endothelial Cells: Early Predictors of Cardiovascular Risk in Offspring? Hypertension 2019; 74:32-34. [PMID: 31154902 DOI: 10.1161/hypertensionaha.119.12652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Jane F Reckelhoff
- From the Departments of Cell and Molecular Biology (J.F.R.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| | - Babbette LaMarca
- Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| | - Vesna D Garovic
- Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.).,Departments of Internal Medicine (V.G.), Mayo Clinic, Rochester, MN.,Hypertension and Nephrology (V.G.), Mayo Clinic, Rochester, MN
| | - Barbara T Alexander
- Physiology and Biophysics (B.T.A.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| |
Collapse
|
44
|
Shah A, Cooke CLM, Kirschenman RD, Quon AL, Morton JS, Care AS, Davidge ST. Sex-specific effects of advanced maternal age on cardiovascular function in aged adult rat offspring. Am J Physiol Heart Circ Physiol 2018; 315:H1724-H1734. [DOI: 10.1152/ajpheart.00375.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pregnancy at an advanced maternal age has an increased risk of complications for both the mothers and their offspring. We have previously shown that advanced maternal age in a rat model leads to poor fetal outcomes, maternal vascular dysfunction, and hypertension, concordant with findings in humans. Moreover, offspring from aged dams had sex-specific cardiovascular dysfunction in young adulthood. However, the detrimental impact of aging on the cardiovascular system of the offspring in this model is unknown. We hypothesized that offspring born to aged dams (9.5–10 mo old) would have impaired cardiovascular function at 12 mo of age. Echocardiographic data revealed signs of mild left ventricular diastolic dysfunction in only male offspring from aged dams [isovolumetric relaxation time: 34.27 ± 2.04 in the young dam group vs. 27.61 ± 0.99 ms in the aged dam group, P < 0.01; mitral annular velocity ratio ( E′/ A′): 1.08 ± 0.04 in the young dam group vs. 0.96 ± 0.02 in the aged dam group, P < 0.05]. We have previously shown that in young adulthood (4 mo of age), male, but not female, offspring born to aged dams had impaired recovery from ischemia-reperfusion injury. Aging did not alter the susceptibility of female offspring to ischemia-reperfusion injury. Interestingly, wire myography data revealed that male offspring from aged dams had enhanced vascular sensitivity to methacholine (negative log of EC50: 7.4 ± 0.08 in young dams vs. 7.9 ± 0.11 in aged dams, P = 0.007) due, in part, to increased prostaglandin-mediated vasodilation. Despite intact endothelium-dependent relaxation, female offspring from aged dams had elevated systolic blood pressure (125.3 ± 4.2 mmHg in young dams vs. 144.0 ± 6.9 mmHg in aged dams, P = 0.03). These data highlight sex-specific mechanisms underlying cardiovascular programming in offspring born to dams of advanced age. NEW & NOTEWORTHY Our study demonstrated that adult male and female offspring (12 mo old) born to aged dams had impaired cardiac diastolic function and increased blood pressure, respectively, signifying sex-specific differential cardiovascular effects of advanced maternal age.
Collapse
Affiliation(s)
- Amin Shah
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Christy-Lynn M. Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Lois Hole Hospital for Women, Edmonton, Alberta, Canada
| | - Raven D. Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Anita L. Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Jude S. Morton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Alison S. Care
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Sandra T. Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
45
|
Kuo AH, Li C, Huber HF, Clarke GD, Nathanielsz PW. Intrauterine growth restriction results in persistent vascular mismatch in adulthood. J Physiol 2018; 596:5777-5790. [PMID: 29098705 PMCID: PMC6265527 DOI: 10.1113/jp275139] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/31/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Intrauterine growth restriction (IUGR) increases offspring risk of chronic diseases later in life, including cardiovascular dysfunction. Our prior studies suggest biventricular cardiac dysfunction and vascular impairment in baboons who were IUGR at birth because of moderate maternal nutrient reduction. The current study reveals changes in artery sizes, distensibility, and blood flow pattern in young adult IUGR baboons, which may contribute to cardiac stress. The pattern of abnormality observed suggests that vascular redistribution seen with IUGR in fetal life may continue into adulthood. ABSTRACT Maternal nutrient reduction induces intrauterine growth restriction (IUGR), increasing risks of chronic diseases later in life, including cardiovascular dysfunction. Using ultrasound, we determined regional blood flow, blood vessel sizes, and distensibility in IUGR baboons (8 males, 8 females, 8.8 years, similar to 35 human years) and controls (12 males, 12 females, 9.5 years). The measured blood vessels were larger in size in the males compared to females before but not after normalization to body surface area. Smaller IUGR normalized blood vessel sizes were observed in the femoral and external iliac arteries but not the brachial or common carotid arteries and not correlated significantly with birth weight. Mild decrease in distensibility in the IUGR group was seen in the iliac but not the carotid arteries without between-sex differences. In IUGR baboons there was increased carotid arterial blood flow velocity during late systole and diastole. Overall, our findings support the conclusion that region specific vascular and haemodynamic changes occur with IUGR, which may contribute to the occurrence of later life cardiac dysfunction. The pattern of alteration observed suggests vascular redistribution efforts in response to challenges in the perinatal period may persist into adulthood. Further studies are needed to determine the life course progression of these changes.
Collapse
Affiliation(s)
- Anderson H. Kuo
- Department of Radiology and Research Imaging InstituteUniversity of Texas Health Science Center at San AntonioSan AntonioTXUSA
| | - Cun Li
- Department of Animal ScienceUniversity of WyomingLaramieWYUSA
- Southwest National Primate Research CenterSan AntonioTXUSA
| | | | - Geoffrey D. Clarke
- Department of Radiology and Research Imaging InstituteUniversity of Texas Health Science Center at San AntonioSan AntonioTXUSA
- Southwest National Primate Research CenterSan AntonioTXUSA
| | - Peter W. Nathanielsz
- Department of Animal ScienceUniversity of WyomingLaramieWYUSA
- Southwest National Primate Research CenterSan AntonioTXUSA
| |
Collapse
|
46
|
Brennan LJ, Goulopoulou S, Bourque SL. Prenatal therapeutics and programming of cardiovascular function. Pharmacol Res 2018; 139:261-272. [PMID: 30458216 DOI: 10.1016/j.phrs.2018.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVD) are a leading cause of mortality worldwide. Despite recognizing the importance of risk factors in dictating CVD susceptibility and onset, patient treatment remains a challenging endeavor. Increasingly, the benefits of prevention and mitigation of risk factors earlier in life are being acknowledged. The developmental origins of health and disease posits that insults during specific periods of development can influence long-term health outcomes; this occurs because the developing organism is highly plastic, and hence vulnerable to environmental perturbations. By extension, targeted therapeutics instituted during critical periods of development may confer long-term protection, and thus reduce the risk of CVD in later life. This review provides a brief overview of models of developmental programming, and then discusses the impact of perinatal therapeutic interventions on long-term cardiovascular function in the offspring. The discussion focuses on bioactive food components, as well as pharmacological agents currently approved for use in pregnancy; in short, those agents most likely to be used in pregnancy and early childhood.
Collapse
Affiliation(s)
- Lesley J Brennan
- Department of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Canada.
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, United States.
| | - Stephane L Bourque
- Department of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Canada.
| |
Collapse
|
47
|
Thompson LP, Chen L, Polster BM, Pinkas G, Song H. Prenatal hypoxia impairs cardiac mitochondrial and ventricular function in guinea pig offspring in a sex-related manner. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1232-R1241. [PMID: 30365351 DOI: 10.1152/ajpregu.00224.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Adverse intrauterine conditions cause fetal growth restriction and increase the risk of adult cardiovascular disease. We hypothesize that intrauterine hypoxia impairs fetal heart function, is sustained after birth, and manifests as both cardiac and mitochondrial dysfunction in offspring guinea pigs (GPs). Pregnant GPs were exposed to 10.5% O2 (HPX) at 50 days of gestation (full term = 65 days) or normoxia (NMX) for the duration of the pregnancy. Pups were allowed to deliver vaginally and raised in a NMX environment. At 90 days of age, mean arterial pressure (MAP) was measured in anesthetized GPs. NMX and prenatally HPX offspring underwent echocardiographic imaging for in vivo measurement of left ventricular cardiac morphology and function, and O2 consumption rates and complex IV enzyme activity were measured from isolated cardiomyocytes and mitochondria, respectively. Prenatal HPX increased ( P < 0.01) MAP (52.3 ± 1.3 and 58.4 ± 1.1 mmHg in NMX and HPX, respectively) and decreased ( P < 0.05) stroke volume (439.8 ± 54.5 and 289.4 ± 15.8 μl in NMX and HPX, respectively), cardiac output (94.4 ± 11.2 and 67.3 ± 3.8 ml/min in NMX and HPX, respectively), ejection fraction, and fractional shortening in male, but not female, GPs. HPX had no effect on left ventricular wall thickness or end-diastolic volume in either sex. HPX reduced mitochondrial maximal respiration and respiratory reserve capacity and complex IV activity rates in hearts of male, but not female, GPs. Prenatal HPX is a programming stimulus that increases MAP and decreases cardiac and mitochondrial function in male offspring. Sex-related differences in the contractile and mitochondrial responses suggest that female GPs are protected from cardiovascular programming of prenatal HPX.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Ling Chen
- Department of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Brian M Polster
- Department of Anesthesiology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Gerard Pinkas
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Hong Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
48
|
Niu Y, Kane AD, Lusby CM, Allison BJ, Chua YY, Kaandorp JJ, Nevin-Dolan R, Ashmore TJ, Blackmore HL, Derks JB, Ozanne SE, Giussani DA. Maternal Allopurinol Prevents Cardiac Dysfunction in Adult Male Offspring Programmed by Chronic Hypoxia During Pregnancy. Hypertension 2018; 72:971-978. [PMID: 30354714 PMCID: PMC6135482 DOI: 10.1161/hypertensionaha.118.11363] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Integrating functional and molecular levels, we investigated the effects of maternal treatment with a xanthine oxidase inhibitor on the programming of cardiac dysfunction in adult offspring using an established rat model of hypoxic pregnancy. Female Wistar rats were divided into normoxic or hypoxic (13% O2) pregnancy±maternal allopurinol treatment (30 mg kg-1 d-1). At 4 months, hearts were isolated from 1 male per litter per outcome variable to determine cardiac function and responses to ischemia-reperfusion in a Langendorff preparation. Sympathetic dominance, perfusate CK (creatine kinase) and LDH (lactate dehydrogenase) and the cardiac protein expression of the β1-adrenergic receptor, the M2 Ach receptor (muscarinic type-2 acetylcholine receptor), and the SERCA2a (sarcoplasmic reticulum Ca2+ ATPase 2a) were determined. Relative to controls, offspring from hypoxic pregnancy showed elevated left ventricular end diastolic pressure (+34.7%), enhanced contractility (dP/dtmax, +41.6%), reduced coronary flow rate (-21%) and an impaired recovery to ischemia-reperfusion (left ventricular diastolic pressure, area under the curve recovery -19.1%; all P<0.05). Increased sympathetic reactivity (heart rate, +755.5%; left ventricular diastolic pressure, +418.9%) contributed to the enhanced myocardial contractility ( P<0.05). Perfusate CK (+431%) and LDH (+251.3%) and the cardiac expression of SERCA2a (+71.4%) were also elevated ( P<0.05), further linking molecular markers of cardiac stress and injury to dysfunction. Maternal allopurinol restored all functional and molecular indices of cardiac pathology. The data support a link between xanthine oxidase-derived oxidative stress in hypoxic pregnancy and cardiac dysfunction in the adult offspring, providing a target for early intervention in the developmental programming of heart disease.
Collapse
Affiliation(s)
- Youguo Niu
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| | - Andrew D. Kane
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Ciara M. Lusby
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Beth J. Allison
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Yi Yi Chua
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Joepe J. Kaandorp
- University Medical Center, Utrecht, the Netherlands (J.J.K., J.B.D.)
| | - Rhiannon Nevin-Dolan
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Thomas J. Ashmore
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
| | - Heather L. Blackmore
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
| | - Jan B. Derks
- University Medical Center, Utrecht, the Netherlands (J.J.K., J.B.D.)
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| | - Dino A. Giussani
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| |
Collapse
|
49
|
Denney JM, Quinn KH. Gestational Diabetes: Underpinning Principles, Surveillance, and Management. Obstet Gynecol Clin North Am 2018; 45:299-314. [PMID: 29747732 DOI: 10.1016/j.ogc.2018.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gestational diabetes mellitus (GDM) is carbohydrate intolerance resulting in hyperglycemia with onset during pregnancy. The article aims to provide clinicians with a working framework to minimize maternal and neonatal morbidity. Landmark historical and recent data are reviewed and presented to provide clinicians with a quick, easy reference for recognition and management of GDM. Data presented tie in insights with underlying pathophysiologic processes leading to GDM. Screening and diagnostic thresholds are discussed along with management upon diagnosis. Good clinical practice regarding screening, diagnosis, and management of GDM effectively reduces risk and improves outcomes of women and fetuses in affected pregnancies.
Collapse
Affiliation(s)
- Jeffrey M Denney
- Department of Obstetrics and Gynecology, Section on Maternal-Fetal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | - Kristen H Quinn
- Department of Obstetrics and Gynecology, Section on Maternal-Fetal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
50
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|