1
|
Ballasy N, Apantaku I, Dean W, Hemberger M. Off to a good start: The importance of the placental exchange surface - Lessons from the mouse. Dev Biol 2024; 517:248-264. [PMID: 39491740 DOI: 10.1016/j.ydbio.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The role of the chorio-allantoic placenta as the critical nutrient- and oxygen-supplying organ to nourish the demands of the fetus has been well recognized. This function relies on the successful establishment of the placental feto-maternal exchange unit, or interhaemal barrier, across which all nutrients as well as waste products must pass to cross from the maternal to the fetal blood circulation, or vice versa, respectively. As a consequence, defects in the establishment of this elaborate interface lead to fetal growth retardation or even embryonic lethality, depending on the severity of the defect. Beyond this essential role, however, it has also emerged that the functionality of the feto-maternal interface dictates the proper development of specific embryonic organs, with tightest links observed to the formation of the heart. In this article, we build on the foundational strength of the mouse as experimental model in which the placental causality of embryonic defects can be genetically proven. We discuss in detail the formation of the interhaemal barrier that makes up the labyrinth layer of the murine placenta, including insights into drivers of its formation and the interdependence of the cell types that make up this essential interface, from in vivo and in vitro data using mouse trophoblast stem cells. We highlight mouse genetic tools that enable the elucidation of cause-effect relationships between defects driven by either the trophoblast cells of the placenta or by embryonic cell types. We specifically emphasize gene knockouts for which a placental causality of embryonic heart defects has been demonstrated. This in-depth perspective provides much-needed insights while highlighting remaining gaps in knowledge that are essential for gaining a better understanding of the multi-facetted roles of the placenta in setting us up for a healthy start in life well beyond nutritional support alone.
Collapse
Affiliation(s)
- Noura Ballasy
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Ifeoluwa Apantaku
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Wendy Dean
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Dept. of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Myriam Hemberger
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
2
|
Zhang S, Lock MC, Tie M, McMillen IC, Botting KJ, Morrison JL. Cardiac programming in the placentally restricted sheep fetus in early gestation. J Physiol 2024; 602:3815-3832. [PMID: 38975864 DOI: 10.1113/jp286702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
Fetal growth restriction (FGR) occurs in 8% of human pregnancies, and the growth restricted newborn is at a greater risk of developing heart disease in later adult life. In sheep, experimental restriction of placental growth (PR) from conception results in FGR, a decrease in cardiomyocyte endowment and an upregulation of pathological hypertrophic signalling in the fetal heart in late gestation. However, there is no change in the expression of markers of cellular proliferation nor in the level of cardiomyocyte apoptosis in the heart of the PR fetus in late gestation. This suggests that FGR arises early in gestation and programs a decrease in cardiomyocyte endowment in early, rather than late, gestation. Here, control and PR fetal sheep were humanely killed at 55 days' gestation (term, 150 days). Fetal body and heart weight were lower in PR compared with control fetuses and there was evidence of sparing of fetal brain growth. While there was no change in the proportion of cardiomyocytes that were proliferating in the early gestation PR heart, there was an increase in measures of apoptosis, and markers of autophagy and pathological hypertrophy in the PR fetal heart. These changes in early gestation highlight that FGR is associated with evidence of early cell death and compensatory hypertrophic responses of cardiomyocytes in the fetal heart. The data suggest that early placental restriction results in a decrease in the pool of proliferative cardiomyocytes in early gestation, which would limit cardiomyocyte endowment in the heart of the PR fetus in late gestation. KEY POINTS: Placental restriction leading to fetal growth restriction (FGR) and chronic fetal hypoxaemia in sheep results in a decrease in cardiomyocyte endowment in late gestation. FGR did not change cardiomyocyte proliferation during early gestation but did result in increased apoptosis and markers of autophagy in the fetal heart, which may result in the decreased endowment of cardiomyocytes observed in late gestation. FGR in early gestation also results in increased hypoxia inducible factor signalling in the fetal heart, which in turn may result in the altered expression of epigenetic regulators, increased expression of insulin-like growth factor 2 and cardiomyocyte hypertrophy during late gestation and after birth.
Collapse
Affiliation(s)
- Song Zhang
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Michelle Tie
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
3
|
Thompson LP, Song H, Hartnett J. Nicotinamide Riboside, an NAD + Precursor, Protects Against Cardiac Mitochondrial Dysfunction in Fetal Guinea Pigs Exposed to Gestational Hypoxia. Reprod Sci 2024; 31:975-986. [PMID: 37957471 PMCID: PMC10959782 DOI: 10.1007/s43032-023-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
Gestational hypoxia inhibits mitochondrial function in the fetal heart and placenta contributing to fetal growth restriction and organ dysfunction. NAD + deficiency may contribute to a metabolic deficit by inhibiting oxidative phosphorylation and ATP synthesis. We tested the effects of nicotinamide riboside (NR), an NAD + precursor, as a treatment for reversing known mitochondrial dysfunction in hypoxic fetal hearts. Pregnant guinea pigs were housed in room air (normoxia) or placed in a hypoxic chamber (10.5%O2) for the last 14 days of gestation (term = 65 days) and administered either water or NR (1.6 mg/ml) in the drinking bottle. Fetuses were excised at term, and NAD + levels of maternal liver, placenta, and fetal heart ventricles were measured. Indices of mitochondrial function (complex IV activity, sirtuin 3 activity, protein acetylation) and ATP synthesis were measured in fetal heart ventricles of NR-treated/untreated normoxic and hypoxic animals. Hypoxia reduced fetal body weight in both sexes (p = 0.01), which was prevented by NR. Hypoxia had no effect on maternal liver NAD + levels but decreased (p = 0.04) placenta NAD + levels, the latter normalized with NR treatment. Hypoxia had no effect on fetal heart NAD + but decreased (p < 0.05) mitochondrial complex IV and sirtuin 3 activities, ATP content, and increased mitochondrial acetylation, which were all normalized with maternal NR. Hypoxia increased (p < 0.05) mitochondrial acetylation in female fetal hearts but had no effect on other mitochondrial indices. We conclude that maternal NR is an effective treatment for normalizing mitochondrial dysfunction and ATP synthesis in the hypoxic fetal heart.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Jamie Hartnett
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
4
|
Drake RR, Louey S, Thornburg KL. Maturation of lipid metabolism in the fetal and newborn sheep heart. Am J Physiol Regul Integr Comp Physiol 2023; 325:R809-R819. [PMID: 37867472 PMCID: PMC11178298 DOI: 10.1152/ajpregu.00122.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
At birth, the fetus experiences a dramatic change in environment that is accompanied by a shift in myocardial fuel preference from lactate and glucose in fetal life to fatty acid oxidation after birth. We hypothesized that fatty acid metabolic machinery would mature during fetal life in preparation for this extreme metabolic transformation at birth. We quantified the pre- (94-day and 135-day gestation, term ∼147 days) and postnatal (5 ± 4 days postnatal) gene expression and protein levels for fatty acid transporters and enzymes in hearts from a precocial species, the sheep. Gene expression of fatty acid translocase (CD36), acyl-CoA synthetase long-chain 1 (ACSL1), carnitine palmitoyltransferase 1 (CPT1), hydroxy-acyl dehydrogenase (HADH), acetyl-CoA acetyltransferase (ACAT1), isocitrate dehydrogenase (IDH), and glycerol phosphate acyltransferase (GPAT) progressively increased through the perinatal period, whereas several genes [fatty acid transport protein 6 (FATP6), acyl-CoA synthetase long chain 3 (ACSL3), long-chain acyl-CoA dehydrogenase (LCAD), very long-chain acyl-CoA dehydrogenase (VLCAD), pyruvate dehydrogenase kinase (PDK4), phosphatidic acid phosphatase (PAP), and diacylglycerol acyltransferase (DGAT)] were stable in fetal hearts and had high expression after birth. Protein expression of CD36 and ACSL1 progressively increased throughout the perinatal period, whereas protein expression of carnitine palmitoyltransferase 1a (fetal isoform) (CPT1a) decreased and carnitine palmitoyltransferase 1b (adult isoform) (CPT1b) remained constitutively expressed. Using fluorescent-tagged long-chain fatty acids (BODIPY-C12), we demonstrated that fetal (125 ± 1 days gestation) cardiomyocytes produce 59% larger lipid droplets (P < 0.05) compared with newborn (8 ± 1 day) cardiomyocytes. These results provide novel insights into the perinatal maturation of cardiac fatty acid metabolism in a precocial species.NEW & NOTEWORTHY This study characterized the previously unknown expression patterns of genes that regulate the metabolism of free fatty acids in the perinatal sheep myocardium. This study shows that the prenatal myocardium prepares for the dramatic switch from carbohydrate metabolism to near complete reliance on free fatty acids postnatally. Fetal and neonatal cardiomyocytes also demonstrate differing lipid storage mechanisms where fetal cardiomyocytes form larger lipid droplets compared with newborn cardiomyocytes.
Collapse
Affiliation(s)
- Rachel R Drake
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Samantha Louey
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Kent L Thornburg
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
5
|
Song H, Thompson LP. Effects of Gestational Hypoxia on PGC1α and Mitochondrial Acetylation in Fetal Guinea Pig Hearts. Reprod Sci 2023; 30:2996-3009. [PMID: 37138147 PMCID: PMC10556133 DOI: 10.1007/s43032-023-01245-5] [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: 02/06/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Chronic intrauterine hypoxia is a significant pregnancy complication impacting fetal heart growth, metabolism, and mitochondrial function, contributing to cardiovascular programming of the offspring. PGC1α (peroxisome proliferator-activated receptor γ co-activator 1α) is the master regulator of mitochondrial biogenesis. We investigated the effects of hypoxia on PGC1α expression following exposure at different gestational ages. Time-mated pregnant guinea pigs were exposed to normoxia (NMX, 21% O2) or hypoxia (HPX, 10.5% O2) at either 25-day (early-onset) or 50-day (late-onset) gestation, and all fetuses were extracted at term (term = ~65-day gestation). Expression of nuclear PGC1α, sirtuin 1 (SIRT1), AMP-activated protein kinase (AMPK), and mitochondrial sirtuin 3 (SIRT3) was measured, along with SIRT3 activity and mitochondrial acetylation of heart ventricles of male and female fetuses. Early-onset hypoxia increased (P<0.05) fetal cardiac nuclear PGC1α and had no effect on mitochondrial acetylation of either growth-restricted males or females. Late-onset hypoxia had either no effect or decreased (P<0.05) PCC1α expression in males and females, respectively, but increased (P<0.05) mitochondrial acetylation in both sexes. Hypoxia had variable effects on expression of SIRT1, AMPK, SIRT3, and SIRT3 activity depending on the sex. The capacity of the fetal heart to respond to hypoxia differs depending on the gestational age of exposure and sex of the fetus. Further, the effects of late-onset hypoxia on fetal heart function impose a greater risk to male than female fetuses, which has implications toward cardiovascular programming effects of the offspring.
Collapse
Affiliation(s)
- Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
6
|
Tang HS, Xiong Y, Li DZ. Fetal Hemoglobin H Hydrops Fetalis: Another Three Case Reports. Hemoglobin 2023:1-3. [PMID: 37247248 DOI: 10.1080/03630269.2023.2215445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 04/10/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023]
Abstract
We report three cases of fetalis hydrops associated with nondeletional α-thalassemia. Two cases were caused by hemoglobin (Hb) H-Quong Sz disease, and one caused by homozygous Hb Constant Spring. Fetal hydrops occurred in the late second trimester in all three cases. Our study indicates that for pregnancies at risk for fetal nondeletional Hb H disease, strict ultrasound follow-up is particularly important. Even without techniques of intrauterine transfusion treatment, early prenatal diagnosis can enable parents to make timely decisions.
Collapse
Affiliation(s)
- Hai-Shen Tang
- Prenatal Diagnosis Unit, Boai Hospital of Zhongshan, Zhongshan, Guangdong, People's Republic of China
| | - Yi Xiong
- Prenatal Diagnosis Unit, Boai Hospital of Zhongshan, Zhongshan, Guangdong, People's Republic of China
| | - Dong-Zhi Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
7
|
Tian L, Jia Z, Yan Y, Jia Q, Shi W, Cui S, Chen H, Han Y, Zhao X, He K. Low-dose of caffeine alleviates high altitude pulmonary edema via regulating mitochondrial quality control process in AT1 cells. Front Pharmacol 2023; 14:1155414. [PMID: 37081967 PMCID: PMC10110878 DOI: 10.3389/fphar.2023.1155414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Backgrounds: High-altitude pulmonary edema (HAPE) is a life-threatening disease without effective drugs. Caffeine is a small molecule compound with antioxidant biological activity used to treat respiratory distress syndrome. However, it is unclear whether caffeine plays a role in alleviating HAPE.Methods: We combined a series of biological experiments and label-free quantitative proteomics analysis to detect the effect of caffeine on treating HAPE and explore its mechanism in vivo and in vitro.Results: Dry and wet weight ratio and HE staining of pulmonary tissues showed that the HAPE model was constructed successfully, and caffeine relieved pulmonary edema. The proteomic results of mice lungs indicated that regulating mitochondria might be the mechanism by which caffeine reduced HAPE. We found that caffeine blocked the reduction of ATP production and oxygen consumption rate, decreased ROS accumulation, and stabilized mitochondrial membrane potential to protect AT1 cells from oxidative stress damage under hypoxia. Caffeine promoted the PINK1/parkin-dependent mitophagy and enhanced mitochondrial fission to maintain the mitochondria quality control process.Conclusion: Low-dose of caffeine alleviated HAPE by promoting PINK1/parkin-dependent mitophagy and mitochondrial fission to control the mitochondria quality. Therefore, caffeine could be a potential treatment for HAPE.
Collapse
Affiliation(s)
- Liuyang Tian
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Yan Yan
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Qian Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Wenjie Shi
- Technical Research Centre for Prevention and Control of Birth Defects, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Saijia Cui
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Huining Chen
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Yang Han
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Xiaojing Zhao
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Kunlun He
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| |
Collapse
|
8
|
Asghari Ahmadabad M, Pourreza N, Ramezanpour S, Baghersalimi A, Enshaei M, Askari M, Alizadeh A, Izadi E, Darbandi B. An analysis of the distribution and spectrum of alpha thalassemia mutations in Rasht City, North of Iran. Front Pediatr 2023; 11:1039148. [PMID: 37033172 PMCID: PMC10073453 DOI: 10.3389/fped.2023.1039148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Background Alpha thalassemia is one of the most common hereditary hemoglobin disorders worldwide, particularly in the Middle East, including Iran. Therefore, determining the spectrum and distribution of alpha thalassemia mutation is a fundamental component of preventive approaches and management strategies. Methods The present study reviews the genetic testing and blood laboratory results of 455 candidates eligible for marriage who were suspected of being thalassemia carriers and on whom genetic testing was performed from 21 March 2013 to 31 December 2020 in Rasht City. Results A total of 114 (25.05%) alpha thalassemia cases were identified. Fifteen different alpha mutations were found. The most common mutation among the study population was -α3.7 deletion in 55 patients (48.24%), followed by Hb Constant Spring (C.S) in 21 patients (18.42%) and poly A2 in 16 (14.03%). Also, most of the patients were silent carriers. The deletion type of mutation was much more common than non-deletion mutations. Conclusion Our study reveals genetic heterogeneity and alpha thalassemia diversity among the Rasht City population. We expect that these findings will help guide premarital screening and genetic counseling, prenatal diagnosis of thalassemia, preventive strategy development, as well as a compilation of the alpha thalassemia catalog in Guilan province.
Collapse
Affiliation(s)
- Mona Asghari Ahmadabad
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Noushin Pourreza
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Setareh Ramezanpour
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Adel Baghersalimi
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mersedeh Enshaei
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Marjan Askari
- Department of Genetic Disorders,Razi Pathobiology and Genetics Laboratory, Rasht, Iran
| | - Amirhossein Alizadeh
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elahe Izadi
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Bahram Darbandi
- Pediatric Department, Pediatric Diseases Research Center, 17 Shahrivar Children's Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Correspondence: Bahram Darbandi
| |
Collapse
|
9
|
Hufnagel A, Grant ID, Aiken CEM. Glucose and oxygen in the early intrauterine environment and their role in developmental abnormalities. Semin Cell Dev Biol 2022; 131:25-34. [PMID: 35410716 DOI: 10.1016/j.semcdb.2022.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 12/14/2022]
Abstract
The early life environment can have profound impacts on the developing conceptus in terms of both growth and morphogenesis. These impacts can manifest in a variety of ways, including congenital fetal anomalies, placental dysfunction with subsequent effects on fetal growth, and adverse perinatal outcomes, or via effects on long-term health outcomes that may not be detected until later childhood or adulthood. Two key examples of environmental influences on early development are explored: maternal hyperglycaemia and gestational hypoxia. These are increasingly common pregnancy exposures worldwide, with potentially profound impacts on population health. We explore what is known regarding the mechanisms by which these environmental exposures can impact early intrauterine development and thus result in adverse outcomes in the immediate, short, and long term.
Collapse
Affiliation(s)
- Antonia Hufnagel
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Imogen D Grant
- Department of Obstetrics and Gynaecology, University of Cambridge, Box 223, The Rosie Hospital and NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge CB2 0SW, UK
| | - Catherine E M Aiken
- Department of Obstetrics and Gynaecology, University of Cambridge, Box 223, The Rosie Hospital and NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge CB2 0SW, UK; University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
10
|
Smith KLM, Swiderska A, Lock MC, Graham L, Iswari W, Choudhary T, Thomas D, Kowash HM, Desforges M, Cottrell EC, Trafford AW, Giussani DA, Galli GLJ. Chronic developmental hypoxia alters mitochondrial oxidative capacity and reactive oxygen species production in the fetal rat heart in a sex-dependent manner. J Pineal Res 2022; 73:e12821. [PMID: 35941749 PMCID: PMC9540814 DOI: 10.1111/jpi.12821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Abstract
Insufficient oxygen supply (hypoxia) during fetal development leads to cardiac remodeling and a predisposition to cardiovascular disease in later life. Previous work has shown hypoxia causes oxidative stress in the fetal heart and alters the activity and expression of mitochondrial proteins in a sex-dependent manner. However, the functional effects of these modifications on mitochondrial respiration remain unknown. Furthermore, while maternal antioxidant treatments are emerging as a promising new strategy to protect the hypoxic fetus, whether these treatments convey similar protection to cardiac mitochondria in the male or female fetus has not been investigated. Therefore, using an established rat model, we measured the sex-dependent effects of gestational hypoxia and maternal melatonin treatment on fetal cardiac mitochondrial respiration, reactive oxygen species (ROS) production, and lipid peroxidation. Pregnant Wistar rats were subjected to normoxia or hypoxia (13% oxygen) during gestational days (GDs) 6-20 (term ~22 days) with or without melatonin treatment (5 µg/ml in maternal drinking water). On GD 20, mitochondrial aerobic respiration and H2 O2 production were measured in fetal heart tissue, together with lipid peroxidation and citrate synthase (CS) activity. Gestational hypoxia reduced maternal body weight gain (p < .01) and increased placental weight (p < .05) but had no effect on fetal weight or litter size. Cardiac mitochondria from male but not female fetuses of hypoxic pregnancy had reduced respiratory capacity at Complex II (CII) (p < .05), and an increase in H2 O2 production/O2 consumption (p < .05) without any changes in lipid peroxidation. CS activity was also unchanged in both sexes. Despite maternal melatonin treatment increasing maternal and fetal plasma melatonin concentration (p < .001), melatonin treatment had no effect on any of the mitochondrial parameters investigated. To conclude, we show that gestational hypoxia leads to ROS generation from the mitochondrial electron transport chain and affects fetal cardiac mitochondrial respiration in a sex-dependent manner. We also show that maternal melatonin treatment had no effect on these relationships, which has implications for the development of future therapies for hypoxic pregnancies.
Collapse
Affiliation(s)
- Kerri L. M. Smith
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Agnieszka Swiderska
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Mitchell C. Lock
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Lucia Graham
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Wulan Iswari
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Tashi Choudhary
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Donna Thomas
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Hager M. Kowash
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Michelle Desforges
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Elizabeth C. Cottrell
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Andrew W. Trafford
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Dino A. Giussani
- Department of Physiology Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Gina L. J. Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
11
|
Carberry CK, Koval LE, Payton A, Hartwell H, Ho Kim Y, Smith GJ, Reif DM, Jaspers I, Ian Gilmour M, Rager JE. Wildfires and extracellular vesicles: Exosomal MicroRNAs as mediators of cross-tissue cardiopulmonary responses to biomass smoke. ENVIRONMENT INTERNATIONAL 2022; 167:107419. [PMID: 35863239 PMCID: PMC9389917 DOI: 10.1016/j.envint.2022.107419] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/23/2022] [Accepted: 07/13/2022] [Indexed: 05/25/2023]
Abstract
INTRODUCTION Wildfires are a threat to public health world-wide that are growing in intensity and prevalence. The biological mechanisms that elicit wildfire-associated toxicity remain largely unknown. The potential involvement of cross-tissue communication via extracellular vesicles (EVs) is a new mechanism that has yet to be evaluated. METHODS Female CD-1 mice were exposed to smoke condensate samples collected from the following biomass burn scenarios: flaming peat; smoldering peat; flaming red oak; and smoldering red oak, representing lab-based simulations of wildfire scenarios. Lung tissue, bronchoalveolar lavage fluid (BALF) samples, peripheral blood, and heart tissues were collected 4 and 24 h post-exposure. Exosome-enriched EVs were isolated from plasma, physically characterized, and profiled for microRNA (miRNA) expression. Pathway-level responses in the lung and heart were evaluated through RNA sequencing and pathway analyses. RESULTS Markers of cardiopulmonary tissue injury and inflammation from BALF samples were significantly altered in response to exposures, with the greatest changes occurring from flaming biomass conditions. Plasma EV miRNAs relevant to cardiovascular disease showed exposure-induced expression alterations, including miR-150, miR-183, miR-223-3p, miR-30b, and miR-378a. Lung and heart mRNAs were identified with differential expression enriched for hypoxia and cell stress-related pathways. Flaming red oak exposure induced the greatest transcriptional response in the heart, a large portion of which were predicted as regulated by plasma EV miRNAs, including miRNAs known to regulate hypoxia-induced cardiovascular injury. Many of these miRNAs had published evidence supporting their transfer across tissues. A follow-up analysis of miR-30b showed that it was increased in expression in the heart of exposed mice in the absence of changes to its precursor molecular, pri-miR-30b, suggesting potential transfer from external sources (e.g., plasma). DISCUSSION This study posits a potential mechanism through which wildfire exposures induce cardiopulmonary responses, highlighting the role of circulating plasma EVs in intercellular and systems-level communication between tissues.
Collapse
Affiliation(s)
- Celeste K Carberry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lauren E Koval
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis Payton
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hadley Hartwell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yong Ho Kim
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
| | - Gregory J Smith
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - David M Reif
- Bioinformatics Research Center, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ilona Jaspers
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA; Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Ian Gilmour
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Julia E Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA; Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Hansell JA, Richter HG, Camm EJ, Herrera EA, Blanco CE, Villamor E, Patey OV, Lock MC, Trafford AW, Galli GLJ, Giussani DA. Maternal melatonin: Effective intervention against developmental programming of cardiovascular dysfunction in adult offspring of complicated pregnancy. J Pineal Res 2022; 72:e12766. [PMID: 34634151 DOI: 10.1111/jpi.12766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/01/2022]
Abstract
Adopting an integrative approach, by combining studies of cardiovascular function with those at cellular and molecular levels, this study investigated whether maternal treatment with melatonin protects against programmed cardiovascular dysfunction in the offspring using an established rodent model of hypoxic pregnancy. Wistar rats were divided into normoxic (N) or hypoxic (H, 10% O2 ) pregnancy ± melatonin (M) treatment (5 μg·ml-1 .day-1 ) in the maternal drinking water. Hypoxia ± melatonin treatment was from day 15-20 of gestation (term is ca. 22 days). To control for possible effects of maternal hypoxia-induced reductions in maternal food intake, additional dams underwent pregnancy under normoxic conditions but were pair-fed (PF) to the daily amount consumed by hypoxic dams from day 15 of gestation. In one cohort of animals from each experimental group (N, NM, H, HM, PF, PFM), measurements were made at the end of gestation. In another, following delivery of the offspring, investigations were made at adulthood. In both fetal and adult offspring, fixed aorta and hearts were studied stereologically and frozen hearts were processed for molecular studies. In adult offspring, mesenteric vessels were isolated and vascular reactivity determined by in-vitro wire myography. Melatonin treatment during normoxic, hypoxic or pair-fed pregnancy elevated circulating plasma melatonin in the pregnant dam and fetus. Relative to normoxic pregnancy, hypoxic pregnancy increased fetal haematocrit, promoted asymmetric fetal growth restriction and resulted in accelerated postnatal catch-up growth. Whilst fetal offspring of hypoxic pregnancy showed aortic wall thickening, adult offspring of hypoxic pregnancy showed dilated cardiomyopathy. Similarly, whilst cardiac protein expression of eNOS was downregulated in the fetal heart, eNOS protein expression was elevated in the heart of adult offspring of hypoxic pregnancy. Adult offspring of hypoxic pregnancy further showed enhanced mesenteric vasoconstrictor reactivity to phenylephrine and the thromboxane mimetic U46619. The effects of hypoxic pregnancy on cardiovascular remodelling and function in the fetal and adult offspring were independent of hypoxia-induced reductions in maternal food intake. Conversely, the effects of hypoxic pregnancy on fetal and postanal growth were similar in pair-fed pregnancies. Whilst maternal treatment of normoxic or pair-fed pregnancies with melatonin on the offspring cardiovascular system was unremarkable, treatment of hypoxic pregnancies with melatonin in doses lower than those recommended for overcoming jet lag in humans enhanced fetal cardiac eNOS expression and prevented all alterations in cardiovascular structure and function in fetal and adult offspring. Therefore, the data support that melatonin is a potential therapeutic target for clinical intervention against developmental origins of cardiovascular dysfunction in pregnancy complicated by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Jeremy A Hansell
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hans G Richter
- Facultad de Medicina, Instituto de Anatomía, Histología y Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Emily J Camm
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emilio A Herrera
- Programa de Fisiopatología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Carlos E Blanco
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Eduardo Villamor
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Olga V Patey
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Mitchell C Lock
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Gina L J Galli
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Dino A Giussani
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge BHF Centre for Research Excellence, Cambridge, UK
- Cambridge Strategic Research Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
13
|
DeFreitas MJ, Katsoufis CP, Benny M, Young K, Kulandavelu S, Ahn H, Sfakianaki A, Abitbol CL. Educational Review: The Impact of Perinatal Oxidative Stress on the Developing Kidney. Front Pediatr 2022; 10:853722. [PMID: 35844742 PMCID: PMC9279889 DOI: 10.3389/fped.2022.853722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/13/2022] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress occurs when there is an imbalance between reactive oxygen species/reactive nitrogen species and antioxidant systems. The interplay between these complex processes is crucial for normal pregnancy and fetal development; however, when oxidative stress predominates, pregnancy related complications and adverse fetal programming such as preterm birth ensues. Understanding how oxidative stress negatively impacts outcomes for the maternal-fetal dyad has allowed for the exploration of antioxidant therapies to prevent and/or mitigate disease progression. In the developing kidney, the negative impact of oxidative stress has also been noted as it relates to the development of hypertension and kidney injury mostly in animal models. Clinical research addressing the implications of oxidative stress in the developing kidney is less developed than that of the neurodevelopmental and respiratory conditions of preterm infants and other vulnerable neonatal groups. Efforts to study the oxidative stress pathway along the continuum of the perinatal period using a team science approach can help to understand the multi-organ dysfunction that the maternal-fetal dyad sustains and guide the investigation of antioxidant therapies to ameliorate the global toxicity. This educational review will provide a comprehensive and multidisciplinary perspective on the impact of oxidative stress during the perinatal period in the development of maternal and fetal/neonatal complications, and implications on developmental programming of accelerated aging and cardiovascular and renal disease for a lifetime.
Collapse
Affiliation(s)
- Marissa J DeFreitas
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States.,Department of Pediatrics, Batchelor Children's Research Institute, University of Miami, Miami, FL, United States
| | - Chryso P Katsoufis
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States.,Department of Pediatrics, Batchelor Children's Research Institute, University of Miami, Miami, FL, United States
| | - Merline Benny
- Department of Pediatrics, Batchelor Children's Research Institute, University of Miami, Miami, FL, United States.,Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL, United States
| | - Karen Young
- Department of Pediatrics, Batchelor Children's Research Institute, University of Miami, Miami, FL, United States.,Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL, United States
| | - Shathiyah Kulandavelu
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States.,Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, United States
| | - Hyunyoung Ahn
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miami, FL, United States
| | - Anna Sfakianaki
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miami, FL, United States
| | - Carolyn L Abitbol
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States.,Department of Pediatrics, Batchelor Children's Research Institute, University of Miami, Miami, FL, United States
| |
Collapse
|
14
|
Song H, Polster BM, Thompson LP. Chronic hypoxia alters cardiac mitochondrial complex protein expression and activity in fetal guinea pigs in a sex-selective manner. Am J Physiol Regul Integr Comp Physiol 2021; 321:R912-R924. [PMID: 34730023 PMCID: PMC8714812 DOI: 10.1152/ajpregu.00004.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
We hypothesize that intrauterine hypoxia (HPX) alters the mitochondrial phenotype in fetal hearts contributing to developmental programming. Pregnant guinea pigs were exposed to normoxia (NMX) or hypoxia (HPX, 10.5% O2), starting at early [25 days (25d), 39d duration] or late gestation (50d, 14d duration). Near-term (64d) male and female fetuses were delivered by hysterotomy from anesthetized sows, and body/organ weights were measured. Left ventricles of fetal hearts were excised and frozen for measurement of expression of complex (I-V) subunits, fusion (Mfn2/OPA1) and fission (DRP1/Fis1) proteins, and enzymatic rates of I and IV from isolated mitochondrial proteins. Chronic HPX decreased fetal body weight and increased relative placenta weight regardless of timing. Early-onset HPX increased I, III, and V subunit levels, increased complex I but decreased IV activities in males but not females (all P < 0.05). Late-onset HPX decreased (P < 0.05) I, III, and V levels in both sexes but increased I and decreased IV activities in males only. Both HPX conditions decreased cardiac mitochondrial DNA content in males only. Neither early- nor late-onset HPX had any effect on Mfn2 levels but increased OPA1 in both sexes. Both HPX treatments increased DRP1/Fis1 levels in males. In females, early-onset HPX increased DRP1 with no effect on Fis1, whereas late-onset HPX increased Fis1 with no effect on DRP1. We conclude that both early- and late-onset HPX disrupts the expression/activities of select complexes that could reduce respiratory efficiency and shifts dynamics toward fission in fetal hearts. Thus, intrauterine HPX disrupts the mitochondrial phenotype predominantly in male fetal hearts, potentially altering cardiac metabolism and predisposing the offspring to heart dysfunction.
Collapse
Affiliation(s)
- Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Loren P Thompson
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, Maryland
| |
Collapse
|
15
|
Abstract
Heart disease remains one of the greatest killers. In addition to genetics and traditional lifestyle risk factors, we now understand that adverse conditions during pregnancy can also increase susceptibility to cardiovascular disease in the offspring. Therefore, the mechanisms by which this occurs and possible preventative therapies are of significant contemporary interest to the cardiovascular community. A common suboptimal pregnancy condition is a sustained reduction in fetal oxygenation. Chronic fetal hypoxia results from any pregnancy with increased placental vascular resistance, such as in preeclampsia, placental infection, or maternal obesity. Chronic fetal hypoxia may also arise during pregnancy at high altitude or because of maternal respiratory disease. This article reviews the short- and long-term effects of hypoxia on the fetal cardiovascular system, and the importance of chronic fetal hypoxia in triggering a developmental origin of future heart disease in the adult progeny. The work summarizes evidence derived from human studies as well as from rodent, avian, and ovine models. There is a focus on the discovery of the molecular link between prenatal hypoxia, oxidative stress, and increased cardiovascular risk in adult offspring. Discussion of mitochondria-targeted antioxidant therapy offers potential targets for clinical intervention in human pregnancy complicated by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Dino A Giussani
- Department of Physiology, Development, and Neuroscience; The Barcroft Centre; Cambridge Cardiovascular British Heart Foundation Centre for Research Excellence; and Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, UK
| |
Collapse
|
16
|
Developmental programming of cardiovascular function: a translational perspective. Clin Sci (Lond) 2021; 134:3023-3046. [PMID: 33231619 DOI: 10.1042/cs20191210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
The developmental origins of health and disease (DOHaD) is a concept linking pre- and early postnatal exposures to environmental influences with long-term health outcomes and susceptibility to disease. It has provided a new perspective on the etiology and evolution of chronic disease risk, and as such is a classic example of a paradigm shift. What first emerged as the 'fetal origins of disease', the evolution of the DOHaD conceptual framework is a storied one in which preclinical studies played an important role. With its potential clinical applications of DOHaD, there is increasing desire to leverage this growing body of preclinical work to improve health outcomes in populations all over the world. In this review, we provide a perspective on the values and limitations of preclinical research, and the challenges that impede its translation. The review focuses largely on the developmental programming of cardiovascular function and begins with a brief discussion on the emergence of the 'Barker hypothesis', and its subsequent evolution into the more-encompassing DOHaD framework. We then discuss some fundamental pathophysiological processes by which developmental programming may occur, and attempt to define these as 'instigator' and 'effector' mechanisms, according to their role in early adversity. We conclude with a brief discussion of some notable challenges that hinder the translation of this preclinical work.
Collapse
|
17
|
Thompson LP, Turan S, Aberdeen GW. Sex differences and the effects of intrauterine hypoxia on growth and in vivo heart function of fetal guinea pigs. Am J Physiol Regul Integr Comp Physiol 2020; 319:R243-R254. [PMID: 32639864 DOI: 10.1152/ajpregu.00249.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We hypothesized that the physiological adaptations of the fetus in response to chronic intrauterine hypoxia depend on its sex and the gestational age of exposure. Pregnant guinea pigs were exposed to room air (normoxia, NMX) or 10.5% O2 (hypoxia, HPX) at either 25 days (early onset) or 50 days (late onset) of gestation until term (~65 days). We evaluated the effects of HPX on hemodynamic and cardiac function indices using Doppler ultrasound and determined sex-related differences in near-term fetuses. Indices of uterine/umbilical artery pulsatility (PI index) and fetal heart systolic and diastolic function [Tei index and passive filling (E-wave) to filling due to atrial contraction (A-wave) (E/A ratios), respectively] were measured in utero and fetal body (FBW) and organ weights measured from extracted fetuses. Both early- and late-onset HPX decreased FBW in both males and females, had no effect on placenta weights, and increased placenta weight-to-FBW ratios. Early- but not late-onset HPX increased uterine artery PI, but neither HPX condition affected umbilical artery PI. Early-onset HPX increased left ventricle E/A ratios in both males and females, whereas late-onset HPX increased the right ventricle E/A ratio in females only. Hypoxia had no effect on the Tei index in either sex. Early- and late-onset HPX induce placental insufficiency and fetal growth restriction and increase diastolic filling depending on the sex, with female fetuses having a greater capacity than males to compensate for intrauterine hypoxia.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shifa Turan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Graham W Aberdeen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Luewan S, Charoenkwan P, Sirichotiyakul S, Tongsong T. Fetal haemoglobin H-Constant Spring disease: a role for intrauterine management. Br J Haematol 2020; 190:e233-e236. [PMID: 32488867 DOI: 10.1111/bjh.16809] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Suchaya Luewan
- Department of Obstetrics and Gynecology, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Theera Tongsong
- Department of Obstetrics and Gynecology, Chiang Mai, Thailand
| |
Collapse
|
19
|
Maki Y, Nygard K, Hammond RR, Regnault TRH, Richardson BS. Maternal Undernourishment in Guinea Pigs Leads to Fetal Growth Restriction with Increased Hypoxic Cells and Oxidative Stress in the Brain. Dev Neurosci 2020; 41:290-299. [PMID: 32316015 DOI: 10.1159/000506939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/28/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We determined whether maternal nutrient restriction (MNR) in guinea pigs leading to fetal growth restriction (FGR) impacts markers for brain hypoxia and oxidative stress. METHODS Guinea pigs were fed ad libitum (control) or 70% of the control diet before pregnancy, switching to 90% at mid-pregnancy (MNR). Near term, hypoxyprobe-1 (HP-1) was injected into pregnant sows. Fetuses were then necropsied and brain tissues were processed for HP-1 (hypoxia marker) and 4HNE, 8-OHdG, and 3-nitrotyrosine (oxidative stress markers) immunoreactivity (IR). RESULTS FGR-MNR fetal and brain weights were decreased 38 and 12%, respectively, with brain/fetal weights thereby increased 45% as a measure of brain sparing, and more so in males than females. FGR-MNR HP-1 IR was increased in most of the brain regions studied, and more so in males than females, while 4HNE and 8-OHdG IR were increased in select brain regions, but with no sex differences. CONCLUSIONS Chronic hypoxia is likely to be an important signaling mechanism in the FGR brain, but with males showing more hypoxia than females. This may involve sex differences in adaptive decreases in growth and normalizing of oxygen, with implications for sex-specific alterations in brain development and risk for later neuropsychiatric disorder.
Collapse
Affiliation(s)
- Yohei Maki
- Department of Obstetrics and Gynecology, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Karen Nygard
- Biotron Integrated Microscopy Facility, University of Western Ontario, London, Ontario, Canada
| | - Robert R Hammond
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada.,Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Timothy R H Regnault
- Department of Obstetrics and Gynecology, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,The Children's Health Research Institute, London, Ontario, Canada
| | - Bryan S Richardson
- Department of Obstetrics and Gynecology, University of Western Ontario, London, Ontario, Canada, .,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada, .,The Children's Health Research Institute, London, Ontario, Canada,
| |
Collapse
|
20
|
Fetal Hypoxia Impacts on Proliferation and Differentiation of Sca-1 + Cardiac Progenitor Cells and Maturation of Cardiomyocytes: A Role of MicroRNA-210. Genes (Basel) 2020; 11:genes11030328. [PMID: 32244901 PMCID: PMC7140790 DOI: 10.3390/genes11030328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is one of the most frequent and severe stresses to an organism’s homeostatic mechanisms, and hypoxia during gestation has profound adverse effects on the heart development increasing the occurrence of congenital heart defects (CHDs). Cardiac progenitor cells (CPCs) are responsible for early heart development and the later occurrence of heart disease. However, the mechanism of how hypoxic stress affects CPC fate decisions and contributes to CHDs remains a topic of debate. Here we examined the effect of hypoxic stress on the regulations of CPC fate decisions and the potential mechanism. We found that experimental induction of hypoxic responses compromised CPC function by regulating CPC proliferation and differentiation and restraining cardiomyocyte maturation. In addition, echocardiography indicated that fetal hypoxia reduced interventricular septum thickness at diastole and the ejection time, but increased the heart rate, in mouse young adult offspring with a gender-related difference. Further study revealed that hypoxia upregulated microRNA-210 expression in Sca-1+ CPCs and impeded the cell differentiation. Blockage of microRNA-210 with LNA-anti-microRNA-210 significantly promoted differentiation of Sca-1+ CPCs into cardiomyocytes. Thus, the present findings provide clear evidence that hypoxia alters CPC fate decisions and reveal a novel mechanism of microRNA-210 in the hypoxic effect, raising the possibility of microRNA-210 as a potential therapeutic target for heart disease.
Collapse
|
21
|
Darby JRT, Varcoe TJ, Orgeig S, Morrison JL. Cardiorespiratory consequences of intrauterine growth restriction: Influence of timing, severity and duration of hypoxaemia. Theriogenology 2020; 150:84-95. [PMID: 32088029 DOI: 10.1016/j.theriogenology.2020.01.080] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/28/2022]
Abstract
At birth, weight of the neonate is used as a marker of the 9-month journey as a fetus. Those neonates born less than the 10th centile for their gestational age are at risk of being intrauterine growth restricted. However, this depends on their genetic potential for growth and the intrauterine environment in which they grew. Alterations in the supply of oxygen and nutrients to the fetus will decrease fetal growth, but these alterations occur due to a range of causes that are maternal, placental or fetal in nature. Consequently, IUGR neonates are a heterogeneous population. For this reason, it is likely that these neonates will respond differently to interventions compared not only to normally grown fetuses, but also to other neonates that are IUGR but have travelled a different path to get there. Thus, a range of models of IUGR should be studied to determine the effects of IUGR on the development and function of the heart and lung and subsequently the impact of interventions to improve development of these organs. Here we focus on a range of models of IUGR caused by manipulation of the maternal, placental or fetal environment on cardiorespiratory outcomes.
Collapse
Affiliation(s)
- Jack R T Darby
- Early Origins of Adult Health Research Group, Australia; School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Tamara J Varcoe
- Early Origins of Adult Health Research Group, Australia; School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Sandra Orgeig
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Australia; School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
22
|
Sirilert S, Charoenkwan P, Sirichotiyakul S, Tongprasert F, Srisupundit K, Luewan S, Tongsong T. Prenatal diagnosis and management of homozygous hemoglobin constant spring disease. J Perinatol 2019; 39:927-933. [PMID: 31097760 DOI: 10.1038/s41372-019-0397-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/21/2019] [Accepted: 04/10/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To describe fetal management of homozygous hemoglobin constant spring (Hb CS). METHODS Six fetuses with homozygous Hb CS disease undergoing intrauterine transfusion (IUT) were comprehensively reviewed. Additionally, when combined with 8 cases previously reported, a total of 14 cases were analyzed. RESULTS The first clues of diagnosis were hydropic changes suggesting fetal anemia. Increased cardiothoracic diameter ratio (CTR) was the most sensitive sonographic marker but slowly changed after IUT, whereas MCA-PSV was the most sensitive in response to IUT. Pre-IUT Hb varied from 1.1% to 6.8%. Gestational age at diagnosis was 17-29 (22.8 ± 3.3) weeks. Rates of adverse obstetric outcomes were relatively high; preterm birth: 35.7%, low birthweight: 42.9%, and fetal growth restriction: 28.6%. All showed good response to IUT with disappearance of hydropic signs and all survived without short-term complications. Their anemia gradually improved in childhood and transfusion independent. CONCLUSION Homozygous Hb CS can cause severe fetal anemia. Early diagnosis and IUT can improve neonatal outcomes, probably preventing adult diseases caused by fetal programming.
Collapse
Affiliation(s)
- Sirinart Sirilert
- Department of Obstetrics and Gynecology, Chiang Mai University, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Fuanglada Tongprasert
- Department of Obstetrics and Gynecology, Chiang Mai University, Chiang Mai, Thailand
| | - Kasemsri Srisupundit
- Department of Obstetrics and Gynecology, Chiang Mai University, Chiang Mai, Thailand
| | - Suchaya Luewan
- Department of Obstetrics and Gynecology, Chiang Mai University, Chiang Mai, Thailand
| | - Theera Tongsong
- Department of Obstetrics and Gynecology, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
23
|
Herrera EA, Ebensperger G, Hernández I, Sanhueza EM, Llanos AJ, Reyes RV. The role of nitric oxide signaling in pulmonary circulation of high- and low-altitude newborn sheep under basal and acute hypoxic conditions. Nitric Oxide 2019; 89:71-80. [PMID: 31063821 DOI: 10.1016/j.niox.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/27/2019] [Accepted: 05/02/2019] [Indexed: 01/05/2023]
Abstract
Nitric oxide (NO) is the main vasodilator agent that drives the rapid decrease of pulmonary vascular resistance for the respiratory onset during the fetal to neonatal transition. Nevertheless, the enhanced NO generation by the neonatal pulmonary arterial endothelium does not prevent development of hypoxic pulmonary hypertension in species without an evolutionary story at high altitude. Therefore, this study aims to describe the limits of the NO function at high-altitude during neonatal life in the sheep as an animal model without tolerance to perinatal hypoxia. We studied the effect of blockade of NO synthesis with l-NAME in the cardiopulmonary response of lowland (580 m) and highland (3600 m) newborn lambs basally and under an episode of acute hypoxia. We also determined the pulmonary expression of proteins that mediate the actions of the NO vasodilator pathway in the pulmonary vasoactive tone and remodeling. We observed an enhanced nitrergic function in highland lambs under basal conditions, evidenced as a markedly greater increase in basal mean pulmonary arterial pressure (mPAP) and resistance (PVR) under blockade of NO synthesis. Further, acute hypoxic challenge in lowland lambs infused with l-NAME markedly increased their mPAP and PVR to values greater than baseline, whilst in highland animals under NO synthesis blockade, these variables did not show additional increase in response to low PO2. Highland animals showed increased pulmonary RhoA expression, decreased PSer188-RhoA fraction, increased PSer311-p65-NFқβ fraction and up-regulated smooth muscle α-actin, relative to lowland controls. Taken together our data suggest that NO-mediated vasodilation is important to keep a low pulmonary vascular resistance under basal conditions and acute hypoxia at low-altitude. At high-altitude, the enhanced nitrergic signaling partially prevents excessive pulmonary hypertension but does not protect against acute hypoxia. The decreased vasodilator efficacy of nitrergic tone in high altitude lambs could be in part due to increased RhoA signaling that opposes to NO action in the hypoxic pulmonary circulation.
Collapse
Affiliation(s)
- Emilio A Herrera
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, RM, Chile
| | - Germán Ebensperger
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, RM, Chile
| | - Ismael Hernández
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile
| | - Emilia M Sanhueza
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile
| | - Aníbal J Llanos
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, RM, Chile
| | - Roberto V Reyes
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, RM, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, RM, Chile.
| |
Collapse
|
24
|
Lv J, Ma Q, Dasgupta C, Xu Z, Zhang L. Antenatal Hypoxia and Programming of Glucocorticoid Receptor Expression in the Adult Rat Heart. Front Physiol 2019; 10:323. [PMID: 31001129 PMCID: PMC6454194 DOI: 10.3389/fphys.2019.00323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.
Collapse
Affiliation(s)
- Juanxiu Lv
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Chiranjib Dasgupta
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
25
|
Shevchenko KM. Morphological features of atrial myocardium embryonic development and its changes caused by hypoxia effect. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mortality and morbidity during the prenatal period of development remain a real problem at the present time. The Scientific Committee EURO-PERISTAT has revealed that mortality of fetuses associated with congenital abnormalities is on average 15–20% across Europe. Hypoxia is one of the top causes of death of fetuses. Since the heart begins to function before birth, influence of teratogenic factors leads to formation of anomalies of its development. Congenital heart defects are the most common of these and occur with a frequency of 24%. Abnormalities associated with the atrium occur with frequency of 6.4 per 10,000 cases. Investigation of structural changes of the atrial myocardium is a key for understanding of pathogenic mechanisms of cardiovascular diseases that are caused by influence of hypoxia. Nowadays, a great deal of research is being dedicated to normal cardiogenesis and much less work is focused on abnormal heart development. There are numerous teratogenic factors such as alcohol, retinoic acid, hyperthermia, hypoxia that are most common causes of heart diseases. The attention of researchers has been predominantly focused on study of changes of the ventricular myocardium under the effect of hypoxia. It is known that the atrium is different from the ventricles by derivation, development and structure. Therefore, the effects of pathological factors on the atrial myocardium will be different as complared to their effect on the ventricles. Also, almost all research has focused on study of consequences of hypoxia at the late stages of cardiogenesis. However, the greatest number of abnormalities is associated with the early embryonic period, as structures that continue development are more sensitive to the effects of harmful factors. Thus, comparative analysis of scientific research devoted to morphological study of atrial myocardium transformations on the cellular and ultrastructural levels under the influence of hypoxia during the stages of cardiogenesis is an important task.
Collapse
|
26
|
Reyes RV, Díaz M, Ebensperger G, Herrera EA, Quezada SA, Hernandez I, Sanhueza EM, Parer JT, Giussani DA, Llanos AJ. The role of nitric oxide in the cardiopulmonary response to hypoxia in highland and lowland newborn llamas. J Physiol 2018; 596:5907-5923. [PMID: 29369354 PMCID: PMC6265547 DOI: 10.1113/jp274340] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/17/2018] [Indexed: 01/19/2023] Open
Abstract
KEY POINTS Perinatal hypoxia causes pulmonary hypertension in neonates, including humans. However, in species adapted to hypoxia, such as the llama, there is protection against pulmonary hypertension. Nitric oxide (NO) is a vasodilatator with an established role in the cardiopulmonary system of many species, but its function in the hypoxic pulmonary vasoconstrictor response in the newborn llama is unknown. Therefore, we studied the role of NO in the cardiopulmonary responses to acute hypoxia in high- and lowland newborn llamas. We show that high- compared to lowland newborn llamas have a reduced pulmonary vasoconstrictor response to acute hypoxia. Protection against excessive pulmonary vasoconstriction in the highland llama is mediated via enhancement of NO pathways, including increased MYPT1 and reduced ROCK expression as well as Ca2+ desensitization. Blunting of pulmonary hypertensive responses to hypoxia through enhanced NO pathways may be an adaptive mechanism to withstand life at high altitude in the newborn llama. ABSTRACT Llamas are born in the Alto Andino with protection against pulmonary hypertension. The physiology underlying protection against pulmonary vasoconstrictor responses to acute hypoxia in highland species is unknown. We determined the role of nitric oxide (NO) in the cardiopulmonary responses to acute hypoxia in high- and lowland newborn llamas. The cardiopulmonary function of newborn llamas born at low (580 m) or high altitude (3600 m) was studied under acute hypoxia, with and without NO blockade. In pulmonary arteries, we measured the reactivity to potassium and sodium nitroprusside (SNP), and in lung we determined the content of cGMP and the expression of the NO-related proteins: BKCa, PDE5, PSer92-PDE5, PKG-1, ROCK1 and 2, MYPT1, PSer695-MYPT1, PThr696-MYPT1, MLC20 and PSer19-MLC20. Pulmonary vascular remodelling was evaluated by morphometry and based on α-actin expression. High- compared to lowland newborn llamas showed lower in vivo pulmonary arterial pressor responses to acute hypoxia. This protection involved enhanced NO function, as NO blockade reverted the effect and the pulmonary arterial dilatator response to SNP was significantly enhanced in highland neonates. The pulmonary expression of ROCK2 and the phosphorylation of MLC20 were lower in high-altitude llamas. Conversely, MYPT1 was up-regulated whilst PSer695-MYPT1 and PThr695-MYPT1 did not change. Enhanced NO-dependent mechanisms were insufficient to prevent pulmonary arterial remodelling. Combined, the data strongly support that in the highland newborn llama reduced ROCK, increased MYPT1 expression and Ca2+ desensitization in pulmonary tissue allow an enhanced NO biology to limit hypoxic pulmonary constrictor responses. Blunting of hypoxic pulmonary hypertensive responses may be an adaptive mechanism to life at high altitude.
Collapse
Affiliation(s)
- Roberto V. Reyes
- Laboratorio de Bioquímica y Biología Molecular de la Hipoxia, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
- International Center for Andean Studies (INCAS)Universidad de ChileSantiagoChile
| | - Marcela Díaz
- Departamento de Promoción de la Salud de la Mujer y el Recién Nacido, Facultad de MedicinaUniversidad de ChileSantiagoChile
- Laboratorio de Mecanismos de Stress y Adaptación Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Germán Ebensperger
- Laboratorio de Mecanismos de Stress y Adaptación Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Emilio A. Herrera
- International Center for Andean Studies (INCAS)Universidad de ChileSantiagoChile
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Sebastián A. Quezada
- Laboratorio de Bioquímica y Biología Molecular de la Hipoxia, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Ismael Hernandez
- Laboratorio de Bioquímica y Biología Molecular de la Hipoxia, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Emilia M. Sanhueza
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Julian T. Parer
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of California San FranciscoCaliforniaUSA
| | - Dino A. Giussani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeUK
| | - Aníbal J. Llanos
- International Center for Andean Studies (INCAS)Universidad de ChileSantiagoChile
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de MedicinaUniversidad de ChileSantiagoChile
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Lv K, Wang G, Shen C, Zhang X, Yao H. Role and mechanism of the nod-like receptor family pyrin domain-containing 3 inflammasome in oral disease. Arch Oral Biol 2018; 97:1-11. [PMID: 30315987 DOI: 10.1016/j.archoralbio.2018.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To summarize evidence and data from experimental studies regarding the role and mechanism of the Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in the pathogenesis of several representative oral diseases. MATERIALS AND METHODS A literature search of PubMed and EBSCO was performed. The literature was searched using a combination of keywords, e.g., NLRP3 inflammasome, inflammation, microorganisms, oral inflammatory diseases, and oral immunological diseases. RESULTS The initiation and activation of the NLRP3 inflammasome are associated with the pathogenesis and progression of several representative oral diseases, including periodontitis, oral lichen planus, dental pulp disease, and oral cavity squamous cell carcinoma. CONCLUSIONS The NLRP3 inflammasome plays a crucial role in the progression of inflammatory and adaptive immune responses. The possible role of the NLRP3 inflammasome in several oral diseases, including not only periodontitis and pulpitis but also mucosal diseases and oral cavity squamous cell carcinoma, may involve the aberrant regulation of inflammatory and immune responses. Understanding the cellular and molecular biology of the NLRP3 inflammasome is necessary because the NLRP3 inflammasome may be a potential therapeutic target for the treatment and prevention of oral inflammatory and immunological diseases.
Collapse
Affiliation(s)
- Kejia Lv
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Guohua Wang
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Chenlu Shen
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Xia Zhang
- Department of Stomatology, Affiliated Yinzhou People Hospital, College of Medicine, Ningbo University, China
| | - Hua Yao
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China.
| |
Collapse
|
30
|
Fajersztajn L, Veras MM. Hypoxia: From Placental Development to Fetal Programming. Birth Defects Res 2018; 109:1377-1385. [PMID: 29105382 DOI: 10.1002/bdr2.1142] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
Hypoxia may influence normal and different pathological processes. Low oxygenation activates a variety of responses, many of them regulated by hypoxia-inducible factor 1 complex, which is mostly involved in cellular control of O2 consumption and delivery, inhibition of growth and development, and promotion of anaerobic metabolism. Hypoxia plays a significant physiological role in fetal development; it is involved in different embryonic processes, for example, placentation, angiogenesis, and hematopoiesis. More recently, fetal hypoxia has been associated directly or indirectly with fetal programming of heart, brain, and kidney function and metabolism in adulthood. In this review, the role of hypoxia in fetal development, placentation, and fetal programming is summarized. Hypoxia is a basic mechanism involved in different pregnancy disorders and fetal health developmental complications. Although there are scientific data showing that hypoxia mediates changes in the growth trajectory of the fetus, modulates gene expression by epigenetic mechanisms, and determines the health status later in adulthood, more mechanistic studies are needed. Furthermore, if we consider that intrauterine hypoxia is not a rare event, and can be a consequence of unavoidable exposures to air pollution, nutritional deficiencies, obesity, and other very common conditions (drug addiction and stress), the health of future generations may be damaged and the incidence of some diseases will markedly increase as a consequence of disturbed fetal programming. Birth Defects Research 109:1377-1385, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lais Fajersztajn
- LIM 05 Departamento de Patologia, Hospital da Clinicas, Faculdade de Medicina Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Mariana Matera Veras
- LIM 05 Departamento de Patologia, Hospital da Clinicas, Faculdade de Medicina Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| |
Collapse
|
31
|
Changes in renal hemodynamics of undernourished fetuses appear earlier than IUGR evidences. J Dev Orig Health Dis 2018; 9:338-343. [PMID: 29374502 DOI: 10.1017/s204017441800003x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study used a sheep model of intrauterine growth restriction, combining maternal undernutrition and twinning, to determine possible markers of early damage to the fetal kidney. The occurrence of early deviations in fetal hemodynamics which may be indicative of changes in blood perfusion was assessed by Doppler ultrasonography. A total of 24 sheep divided in two groups were fed with the same standard grain-based diet but fulfilling either their daily maintenance requirements for pregnancy (control group; n=12, six singleton and six twin pregnancies) or only the 50% of such quantity (food-restricted group; n=12; four singleton and eight twin pregnancies). All the fetuses were assessed by both B-mode and Doppler ultrasonography at Day 115 of pregnancy. Fetal blood supply was affected by maternal undernutrition, although there were still no evidences of brain-sparing excepting in fetuses at greatest challenge (twins in underfed pregnancies). However, there were early changes in the blood supply to the kidneys of underfed fetuses and underfed twins evidenced decreases in kidney size.
Collapse
|
32
|
Lin J, Huang H, Lv G, Xu X, Lin W, Xu X, Cheng J, Zheng M. Chronic prenatal hypoxia impairs cochlear development, a mechanism involving connexin26 expression and promoter methylation. Int J Mol Med 2017; 41:852-858. [PMID: 29207085 PMCID: PMC5752184 DOI: 10.3892/ijmm.2017.3303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022] Open
Abstract
Chronic prenatal hypoxia is a damaging to fetal development and may have various consequences, including hearing loss. Connexin 26 (Cx26) is one of the major protein subunits required for gap junction formation, and has an important role in maintaining homeostasis in the cochlea and normal hearing. Cx26 mutation and expression abnormality are closely associated with inherited nonsyndromic deafness, but the association between Cx26 and prenatal hypoxia is less established. The present study aimed to examine Cx26 expression and aberrant methylation the Cx26 promoter region in the cochlea from rats exposed to chronic prenatal hypoxia. Hematoxylin and eosin staining demonstrated that the number of hair cells in the organ of Corti were less in the hypoxia group. Reverse transcription-quantitative polymerase chain reaction and western blot analysis revealed that protein and mRNA levels of Cx26 were decreased in the hypoxia group compared with the control group. Further bisulfite sequencing analysis revealed that prenatal hypoxia significantly increased the methylation status of the promoter region of the Cx26 gene. These results demonstrate that chronic prenatal hypoxia caused hearing impairment, and suggest that promoter region hypermethylation and expression downregulation of Cx26 underlie the mechanism of action.
Collapse
Affiliation(s)
- Jingcang Lin
- Department of Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Huang Huang
- Department of Histology and Embryology, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Guorong Lv
- Department of Medical Imaging, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Xiangyang Xu
- Department of Histology and Embryology, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Wendong Lin
- Department of Anatomy, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Xianyan Xu
- Department of Anatomy, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Jing Cheng
- Department of Histology and Embryology, Quanzhou Medical College, Quanzhou, Fujian 362100, P.R. China
| | - Ming Zheng
- Department of Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| |
Collapse
|
33
|
Turan S, Aberdeen GW, Thompson LP. Chronic hypoxia alters maternal uterine and fetal hemodynamics in the full-term pregnant guinea pig. Am J Physiol Regul Integr Comp Physiol 2017; 313:R330-R339. [PMID: 28679680 DOI: 10.1152/ajpregu.00056.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/22/2017] [Accepted: 06/28/2017] [Indexed: 12/17/2022]
Abstract
Placental hypoxia is associated with maternal hypertension, placental insufficiency, and fetal growth restriction. In the pregnant guinea pig, prenatal hypoxia during early gestation inhibits cytotrophoblast invasion of spiral arteries, increases maternal blood pressure, and induces fetal growth restriction. In this study the impact of chronic maternal hypoxia on fetal heart structure was evaluated using four-dimensional echocardiography with spatiotemporal image correlation and tomographic ultrasound, and uterine and umbilical artery resistance/pulsatility indexes and fetal heart function were evaluated using pulsed-wave Doppler ultrasound. Pregnant guinea pigs were exposed to normoxia (n = 7) or hypoxia (10.5% O2, n = 9) at 28-30 days gestation, which was maintained until full term (65 days). At full term, fetal heart structure and outflow tracts were evaluated in the four-chamber view. Fetal heart diastolic function was assessed by E wave-to-A wave diastolic filling ratios (E/A ratios) of both ventricles and systolic function by the myocardial performance index (or Tie) of left ventricles of normoxic (n = 21) and hypoxic (n = 17) fetuses. There were no structural abnormalities in fetal hearts. However, hypoxia induced asymmetric fetal growth restriction and increased the placental/fetal weight compared with normoxic controls. Hypoxia increased Doppler resistance and pulsatility indexes in the uterine, but not umbilical, arteries, had no effect on the Tie index, and increased the E/A ratio in left, but not right, ventricles. Thus, prolonged hypoxia, starting at midgestation, increases uterine artery resistance and generates fetal growth restriction at full term. Furthermore, the enhanced cardiac diastolic filling with no changes in systolic function or umbilical artery resistance suggests that the fetal guinea pig systemic circulation undergoes a compensated, adaptive response to prolonged hypoxia exposure.
Collapse
Affiliation(s)
- Sifa Turan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Graham W Aberdeen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Loren P Thompson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Li X, Zhang M, Pan X, Xu Z, Sun M. “Three Hits” Hypothesis for Developmental Origins of Health and Diseases in View of Cardiovascular Abnormalities. Birth Defects Res 2017; 109:744-757. [PMID: 28509412 DOI: 10.1002/bdr2.1037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Xiang Li
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Mengshu Zhang
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences
- Key Laboratory of Biochip Technology in Guangdong province; Southern Medical University; Guangzhou China
- Department of Genetics; Yale University School of Medicine; New Haven Connecticut
| | - Zhice Xu
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Miao Sun
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| |
Collapse
|
35
|
Abstract
Epidemiological evidence links an individual's susceptibility to chronic disease in adult life to events during their intrauterine phase of development. Biologically this should not be unexpected, for organ systems are at their most plastic when progenitor cells are proliferating and differentiating. Influences operating at this time can permanently affect their structure and functional capacity, and the activity of enzyme systems and endocrine axes. It is now appreciated that such effects lay the foundations for a diverse array of diseases that become manifest many years later, often in response to secondary environmental stressors. Fetal development is underpinned by the placenta, the organ that forms the interface between the fetus and its mother. All nutrients and oxygen reaching the fetus must pass through this organ. The placenta also has major endocrine functions, orchestrating maternal adaptations to pregnancy and mobilizing resources for fetal use. In addition, it acts as a selective barrier, creating a protective milieu by minimizing exposure of the fetus to maternal hormones, such as glucocorticoids, xenobiotics, pathogens, and parasites. The placenta shows a remarkable capacity to adapt to adverse environmental cues and lessen their impact on the fetus. However, if placental function is impaired, or its capacity to adapt is exceeded, then fetal development may be compromised. Here, we explore the complex relationships between the placental phenotype and developmental programming of chronic disease in the offspring. Ensuring optimal placentation offers a new approach to the prevention of disorders such as cardiovascular disease, diabetes, and obesity, which are reaching epidemic proportions.
Collapse
Affiliation(s)
- Graham J Burton
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Abigail L Fowden
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Kent L Thornburg
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
36
|
Zou T, Chen D, Yang Q, Wang B, Zhu MJ, Nathanielsz PW, Du M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J Physiol 2017; 595:1547-1562. [PMID: 27891610 DOI: 10.1113/jp273478] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/15/2016] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS Maternal high-fat diet impairs brown adipocyte function and correlates with obesity in offspring. Maternal resveratrol administration recovers metabolic activity of offspring brown adipose tissue. Maternal resveratrol promotes beige adipocyte development in offspring white adipose tissue. Maternal resveratrol intervention protects offspring against high-fat diet-induced obesity. ABSTRACT Promoting beige/brite adipogenesis and thermogenic activity is considered as a promising therapeutic approach to reduce obesity and metabolic syndrome. Maternal obesity impairs offspring brown adipocyte function and correlates with obesity in offspring. We previously found that dietary resveratrol (RES) induces beige adipocyte formation in adult mice. Here, we evaluated further the effect of resveratrol supplementation of pregnant mice on offspring thermogenesis and energy expenditure. Female C57BL/6 J mice were fed a control diet (CON) or a high-fat diet (HFD) with or without 0.2% (w/w) RES during pregnancy and lactation. Male offspring were weaned onto a HFD and maintained on this diet for 11 weeks. The offspring thermogenesis and related regulatory factors in adipose tissue were evaluated. At weaning, HFD offspring had lower thermogenesis in brown and white adipose tissues compared with CON offspring, which was recovered by maternal RES supplementation, along with the appearance of multilocular brown/beige adipocytes and elevated thermogenic gene expression. Adult offspring of RES-treated mothers showed increased energy expenditure and insulin sensitivity when on an obesogenic diet compared with HFD offspring. The elevated metabolic activity was correlated with enhanced brown adipose function and white adipose tissue browning in HFD+RES compared with HFD offspring. In conclusion, RES supplementation of HFD-fed dams during pregnancy and lactation promoted white adipose browning and thermogenesis in offspring at weaning accompanied by persistent beneficial effects in protecting against HFD-induced obesity and metabolic disorders.
Collapse
Affiliation(s)
- Tiande Zou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.,Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qiyuan Yang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Bo Wang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Peter W Nathanielsz
- Wyoming Pregnancy and Life Course Health Centre, Department of Animal Science, University of Wyoming, Laramie, WY, 82071, USA
| | - Min Du
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA.,Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, 100194, China
| |
Collapse
|
37
|
Tarín JJ, García-Pérez MA, Cano A. Obstetric and offspring risks of women's morbid conditions linked to prior anticancer treatments. Reprod Biol Endocrinol 2016; 14:37. [PMID: 27386839 PMCID: PMC4936115 DOI: 10.1186/s12958-016-0169-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 06/16/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Literature shows the effects of type of cancer and/or anticancer treatment on live birth percentages and/or pregnancy and neonatal complications in female cancer survivors. However, studies analyzing the obstetric and offspring risks of the morbid conditions associated with previous anti-cancer treatments are missing. The present review aims to uncover these risks. METHODS A literature search based on publications up to March 2016 identified by PubMed and references cited in relevant articles. RESULTS The morbid conditions associated with prior anticancer treatments including chemotherapy, radiotherapy, surgery, and/or hematopoietic stem-cell transplant may induce not only obstetric and neonatal complications but also long-term effects on offspring. Whereas some risks are predominantly evidenced in untreated women others are observed in both treated and untreated women. These risks may be superimposed on those induced by the current women's trend in Western societies to postpone maternity. CONCLUSIONS Medical professionals should be aware and inform female cancer survivors wishing to have a child not only of the short- and long-term risks to themselves and their prospective offspring of previous anticancer treatments, fertility-preservation technologies, and pregnancy itself, but also of those risks linked to the morbid conditions induced by prior anticancer treatments. Once female cancer survivors wishing to have a child have been properly informed about the risks of reproduction, they will be best placed to make decisions of whether or not to have a biological or donor-conceived child. In addition, when medical professionals be aware of these risks, they will be also best placed to provide appropriate treatments before/during pregnancy in order to prevent or alleviate the impact of these morbid conditions on maternal and offspring health.
Collapse
Affiliation(s)
- Juan J. Tarín
- Department of Cellular Biology, Functional Biology and Physical Anthropology, Faculty of Biological Sciences, University of Valencia, Burjassot, Valencia, 46100 Spain
| | - Miguel A. García-Pérez
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Burjassot, Valencia, 46100 Spain
- Research Unit-INCLIVA, Hospital Clínico de Valencia, Valencia, 46010 Spain
| | - Antonio Cano
- Department of Pediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, 46010 Spain
- Service of Obstetrics and Gynecology, University Clinic Hospital, Valencia, 46010 Spain
| |
Collapse
|
38
|
Intrauterine growth restriction: impact on cardiovascular development and function throughout infancy. Pediatr Res 2016; 79:821-30. [PMID: 26866903 DOI: 10.1038/pr.2016.24] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/05/2015] [Indexed: 01/08/2023]
Abstract
Intrauterine growth restriction (IUGR) refers to the situation where a fetus does not grow according to its genetic growth potential. One of the main causes of IUGR is uteroplacental vascular insufficiency. Under these circumstances of chronic oxygen and nutrient deprivation, the growth-restricted fetus often displays typical circulatory changes, which in part represent adaptations to the suboptimal intrauterine environment. These fetal adaptations aim to preserve oxygen and nutrient supply to vital organs such as the brain, the heart, and the adrenals. These prenatal circulatory adaptations are thought to lead to an altered development of the cardiovascular system and "program" the fetus for life long cardiovascular morbidities. In this review, we discuss the alterations to cardiovascular structure, function, and control that have been observed in growth-restricted fetuses, neonates, and infants following uteroplacental vascular insufficiency. We also discuss the current knowledge on early life surveillance and interventions to prevent progression into chronic disease.
Collapse
|
39
|
Sferruzzi-Perri AN, Camm EJ. The Programming Power of the Placenta. Front Physiol 2016; 7:33. [PMID: 27014074 PMCID: PMC4789467 DOI: 10.3389/fphys.2016.00033] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/25/2016] [Indexed: 12/23/2022] Open
Abstract
Size at birth is a critical determinant of life expectancy, and is dependent primarily on the placental supply of nutrients. However, the placenta is not just a passive organ for the materno-fetal transfer of nutrients and oxygen. Studies show that the placenta can adapt morphologically and functionally to optimize substrate supply, and thus fetal growth, under adverse intrauterine conditions. These adaptations help meet the fetal drive for growth, and their effectiveness will determine the amount and relative proportions of specific metabolic substrates supplied to the fetus at different stages of development. This flow of nutrients will ultimately program physiological systems at the gene, cell, tissue, organ, and system levels, and inadequacies can cause permanent structural and functional changes that lead to overt disease, particularly with increasing age. This review examines the environmental regulation of the placental phenotype with particular emphasis on the impact of maternal nutritional challenges and oxygen scarcity in mice, rats and guinea pigs. It also focuses on the effects of such conditions on fetal growth and the developmental programming of disease postnatally. A challenge for future research is to link placental structure and function with clinical phenotypes in the offspring.
Collapse
Affiliation(s)
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| |
Collapse
|
40
|
Allison BJ, Kaandorp JJ, Kane AD, Camm EJ, Lusby C, Cross CM, Nevin-Dolan R, Thakor AS, Derks JB, Tarry-Adkins JL, Ozanne SE, Giussani DA. Divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease. FASEB J 2016; 30:1968-75. [PMID: 26932929 PMCID: PMC5036970 DOI: 10.1096/fj.201500057] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/26/2016] [Indexed: 12/24/2022]
Abstract
Aging and developmental programming are both associated with oxidative stress and endothelial dysfunction, suggesting common mechanistic origins. However, their interrelationship has been little explored. In a rodent model of programmed cardiovascular dysfunction we determined endothelial function and vascular telomere length in young (4 mo) and aged (15 mo) adult offspring of normoxic or hypoxic pregnancy with or without maternal antioxidant treatment. We show loss of endothelial function [maximal arterial relaxation to acetylcholine (71 ± 3 vs. 55 ± 3%) and increased vascular short telomere abundance (4.2–1.3 kb) 43.0 ± 1.5 vs. 55.1 ± 3.8%) in aged vs. young offspring of normoxic pregnancy (P < 0.05). Hypoxic pregnancy in young offspring accelerated endothelial dysfunction (maximal arterial relaxation to acetylcholine: 42 ± 1%, P < 0.05) but this was dissociated from increased vascular short telomere length abundance. Maternal allopurinol rescued maximal arterial relaxation to acetylcholine in aged offspring of normoxic or hypoxic pregnancy but not in young offspring of hypoxic pregnancy. Aged offspring of hypoxic allopurinol pregnancy compared with aged offspring of untreated hypoxic pregnancy had lower levels of short telomeres (vascular short telomere length abundance 35.1 ± 2.5 vs. 48.2 ± 2.6%) and of plasma proinflammatory chemokine (24.6 ± 2.8 vs. 36.8 ± 5.5 pg/ml, P < 0.05). These data provide evidence for divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease, and aging being decelerated by antioxidants even prior to birth.—Allison, B. J., Kaandorp, J. J., Kane, A. D., Camm, E. J., Lusby, C., Cross, C. M., Nevin-Dolan, R., Thakor, A. S., Derks, J. B., Tarry-Adkins, J. L., Ozanne, S. E., Giussani, D. A. Divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease.
Collapse
Affiliation(s)
- Beth J Allison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Joepe J Kaandorp
- Perinatology, University Medical Center, Utrecht, The Netherlands; and
| | - Andrew D Kane
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Emily J Camm
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ciara Lusby
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Christine M Cross
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rhianon Nevin-Dolan
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Avnesh S Thakor
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jan B Derks
- Perinatology, University Medical Center, Utrecht, The Netherlands; and
| | - Jane L Tarry-Adkins
- Metabolic Research Laboratories and Medical Reseach Council (MRC) Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Susan E Ozanne
- Metabolic Research Laboratories and Medical Reseach Council (MRC) Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Dino A Giussani
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom;
| |
Collapse
|
41
|
Vega CC, Reyes-Castro LA, Rodríguez-González GL, Bautista CJ, Vázquez-Martínez M, Larrea F, Chamorro-Cevallos GA, Nathanielsz PW, Zambrano E. Resveratrol partially prevents oxidative stress and metabolic dysfunction in pregnant rats fed a low protein diet and their offspring. J Physiol 2016; 594:1483-99. [PMID: 26662841 DOI: 10.1113/jp271543] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/04/2015] [Indexed: 12/14/2022] Open
Abstract
Protein restriction in pregnancy produces maternal and offspring metabolic dysfunction potentially as a result of oxidative stress. Data are lacking on the effects of inhibition of oxidative stress. We hypothesized that maternal resveratrol administration decreases oxidative stress, preventing, at least partially, maternal low protein-induced maternal and offspring metabolic dysfunction. In the present study, pregnant wistar rats ate control (C) (20% casein) or a protein-restricted (R) (10% casein) isocaloric diet. Half of each group received resveratrol orally, 20 mg kg(-1) day(-1), throughout pregnancy. Post-delivery, mothers and offspring ate C. Oxidative stress biomarkers and anti-oxidant enzymes were measured in placenta, maternal and fetal liver, and maternal serum corticosterone at 19 days of gestation (dG). Maternal (19 dG) and offspring (postnatal day 110) glucose, insulin, triglycerides, cholesterol, fat and leptin were determined. R mothers showed metabolic dysfunction, increased corticosterone and oxidative stress and reduced anti-oxidant enzyme activity vs. C. R placental and fetal liver oxidative stress biomarkers and anti-oxidant enzyme activity increased. R offspring showed higher male and female leptin, insulin and corticosterone, male triglycerides and female fat than C. Resveratrol decreased maternal leptin and improved maternal, fetal and placental oxidative stress markers. R induced offspring insulin and leptin increases were prevented and other R changes were offspring sex-dependent. Resveratrol partially prevents low protein diet-induced maternal, placental and sex-specific offspring oxidative stress and metabolic dysfunction. Oxidative stress is one mechanism programming offspring metabolic outcomes. These studies provide mechanistic evidence to guide human pregnancy interventions when fetal nutrition is impaired by poor maternal nutrition or placental function.
Collapse
Affiliation(s)
- Claudia C Vega
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Pharmacy Department, National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luis A Reyes-Castro
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Claudia J Bautista
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Magaly Vázquez-Martínez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Fernando Larrea
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Germán A Chamorro-Cevallos
- Pharmacy Department, National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Peter W Nathanielsz
- Wyoming Center for Pregnancy and Life Course Health Research, Department of Animal Science, University Wyoming, Laramie, WY
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
42
|
Zhu X, Gao Q, Tu Q, Zhong Y, Zhu D, Mao C, Xu Z. Prenatal hypoxia enhanced angiotensin II-mediated vasoconstriction via increased oxidative signaling in fetal rats. Reprod Toxicol 2016; 60:21-8. [PMID: 26796766 DOI: 10.1016/j.reprotox.2016.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/29/2015] [Accepted: 01/08/2016] [Indexed: 02/07/2023]
Abstract
Toxic factors could cause in utero hypoxia, and prenatal hypoxia (PH) increased incidence of cardiovascular diseases in late life. It is unclear whether/how PH causes vascular injury during fetal life. This study found that PH significantly increased angiotensin II (Ang II)-mediated vessel contractions in fetal thoracic aortas, which was blocked by losartan, not PD123319, indicating that AT1 receptors played a dominant role in the enhanced fetal vasoconstriction following hypoxia. Prenatal hypoxia increased superoxide production and decreased superoxide dismutase (SOD) expression, associated with the enhanced NADPH oxidase (Nox) 4, but not Nox1 or Nox2 in fetal aortas. Ang II-increased vasoconstriction was inhibited by Nox inhibitor apocynin and SOD mimetic blocker tempol. These findings suggested that PH resulted in Ang II/AT1R-mediated fetal vascular hypertensive re-activity via pathways of Nox4-dependent oxidative stress, providing new information regarding the impact of PH on the functional and molecular development of fetal vascular systems.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qing Tu
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Zhong
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Di Zhu
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology and Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China; Center for Perinatal Biology, Loma Linda University, CA, USA.
| |
Collapse
|
43
|
Zhang P, Zhu D, Chen X, Li Y, Li N, Gao Q, Li L, Zhou X, Lv J, Sun M, Mao C, Xu Z. Prenatal hypoxia promotes atherosclerosis via vascular inflammation in the offspring rats. Atherosclerosis 2015; 245:28-34. [PMID: 26691907 DOI: 10.1016/j.atherosclerosis.2015.11.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/06/2015] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypoxia is a critical contributor to increased risks of cardiovascular diseases, including atherosclerosis, but the detailed mechanism that hypoxia leads to atherosclerosis remains unknown. METHODS Pregnant rats were treated with hypoxia (10.5% oxygen) during pregnancy, and HUVEC cells treated with 1% of oxygen. Blood lipids were tested at fetal stage and adult stage of offspring rats; the level of pro-inflammatory cytokines of HUVEC and offspring rats were investigated, and HIF-1α and NFκB mRNA level were also measured by Q-PCR and Elisa. RESULTS We found that TC, LDL-C, ox-LDL-C, and the receptors of ox-LDL-C (lox-1) of the adult offspring were significantly higher than that of the control, while HDL-C was significantly reduced in hypoxia group. The internal elastic lamina was blocked by smooth muscle cells; and the migration of smooth muscle cells into the intima were observed in hypoxia offspring. Luciferase reporter gene experiment showed that HIF-1α activated NFκB transcription at four discrete binding sites of NFκBp65 promoter, although there was no obvious difference among the four discrete binding sites. Using transfection of pCDNA3.1-HIF-1α on HUVEC cells, HIF-1α significantly activated NFκB transcription at hypoxic conditions (1% O2), and concurrent with increased expression of IL-1β and TNF-α. CONCLUSION Hypoxia during pregnancy activated NFκB transcription to induce pro-inflammatory cytokines, leading to the early stage of atherosclerosis.
Collapse
Affiliation(s)
- Pengjie Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Di Zhu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xionghui Chen
- Emergency Department, First Hospital of Soochow University, Suzhou, China
| | - Yongmei Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Juanxiu Lv
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China; Center for Prenatal Biology, Loma Linda University, CA, USA.
| |
Collapse
|
44
|
Wearing OH, Eme J, Rhen T, Crossley DA. Phenotypic plasticity in the common snapping turtle (Chelydra serpentina): long-term physiological effects of chronic hypoxia during embryonic development. Am J Physiol Regul Integr Comp Physiol 2015; 310:R176-84. [PMID: 26608655 DOI: 10.1152/ajpregu.00293.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/24/2015] [Indexed: 11/22/2022]
Abstract
Studies of embryonic and hatchling reptiles have revealed marked plasticity in morphology, metabolism, and cardiovascular function following chronic hypoxic incubation. However, the long-term effects of chronic hypoxia have not yet been investigated in these animals. The aim of this study was to determine growth and postprandial O2 consumption (V̇o2), heart rate (fH), and mean arterial pressure (Pm, in kPa) of common snapping turtles (Chelydra serpentina) that were incubated as embryos in chronic hypoxia (10% O2, H10) or normoxia (21% O2, N21). We hypothesized that hypoxic development would modify posthatching body mass, metabolic rate, and cardiovascular physiology in juvenile snapping turtles. Yearling H10 turtles were significantly smaller than yearling N21 turtles, both of which were raised posthatching in normoxic, common garden conditions. Measurement of postprandial cardiovascular parameters and O2 consumption were conducted in size-matched three-year-old H10 and N21 turtles. Both before and 12 h after feeding, H10 turtles had a significantly lower fH compared with N21 turtles. In addition, V̇o2 was significantly elevated in H10 animals compared with N21 animals 12 h after feeding, and peak postprandial V̇o2 occurred earlier in H10 animals. Pm of three-year-old turtles was not affected by feeding or hypoxic embryonic incubation. Our findings demonstrate that physiological impacts of developmental hypoxia on embryonic reptiles continue into juvenile life.
Collapse
Affiliation(s)
- Oliver H Wearing
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - John Eme
- Department of Biological Sciences, California State University San Marcos, San Marcos, California
| | - Turk Rhen
- Department of Biology, University of North Dakota, Grand Forks, North Dakota; and
| | - Dane A Crossley
- Department of Biological Sciences, University of North Texas, Denton, Texas
| |
Collapse
|
45
|
Farías JG, Herrera EA, Carrasco-Pozo C, Sotomayor-Zárate R, Cruz G, Morales P, Castillo RL. Pharmacological models and approaches for pathophysiological conditions associated with hypoxia and oxidative stress. Pharmacol Ther 2015; 158:1-23. [PMID: 26617218 DOI: 10.1016/j.pharmthera.2015.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypoxia is the failure of oxygenation at the tissue level, where the reduced oxygen delivered is not enough to satisfy tissue demands. Metabolic depression is the physiological adaptation associated with reduced oxygen consumption, which evidently does not cause any harm to organs that are exposed to acute and short hypoxic insults. Oxidative stress (OS) refers to the imbalance between the generation of reactive oxygen species (ROS) and the ability of endogenous antioxidant systems to scavenge ROS, where ROS overwhelms the antioxidant capacity. Oxidative stress plays a crucial role in the pathogenesis of diseases related to hypoxia during intrauterine development and postnatal life. Thus, excessive ROS are implicated in the irreversible damage to cell membranes, DNA, and other cellular structures by oxidizing lipids, proteins, and nucleic acids. Here, we describe several pathophysiological conditions and in vivo and ex vivo models developed for the study of hypoxic and oxidative stress injury. We reviewed existing literature on the responses to hypoxia and oxidative stress of the cardiovascular, renal, reproductive, and central nervous systems, and discussed paradigms of chronic and intermittent hypobaric hypoxia. This systematic review is a critical analysis of the advantages in the application of some experimental strategies and their contributions leading to novel pharmacological therapies.
Collapse
Affiliation(s)
- Jorge G Farías
- Facultad de Ingeniería y Ciencias, Departamento de Ingeniería Química, Universidad de la Frontera, Casilla 54-D, Temuco, Chile
| | - Emilio A Herrera
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Chile
| | | | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Paola Morales
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Chile
| | - Rodrigo L Castillo
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Chile.
| |
Collapse
|
46
|
Parraguez VH, Mamani S, Cofré E, Castellaro G, Urquieta B, De los Reyes M, Astiz S, Gonzalez-Bulnes A. Disturbances in Maternal Steroidogenesis and Appearance of Intrauterine Growth Retardation at High-Altitude Environments Are Established from Early Pregnancy. Effects of Treatment with Antioxidant Vitamins. PLoS One 2015; 10:e0140902. [PMID: 26560325 PMCID: PMC4641609 DOI: 10.1371/journal.pone.0140902] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/01/2015] [Indexed: 11/19/2022] Open
Abstract
Pregnancies at high-altitudes are influenced by hypoxia and oxidative stress and frequently affected by IUGR. However, a common thought is that early pregnant women visiting altitude have no major complications for gestation development, since IUGR is developed during the second half of pregnancy. Thus, using a well-characterized sheep-model, we aimed to determine whether long- and/or short-term exposure to high-altitude may affect maternal steroidogenesis and therefore embryo-fetal growth from conception. The second aim was to differentiate the relative role of hypoxia and oxidative stress by assessing the effects of supplementation with antioxidant agents during this early-pregnancy stage, which were previously found to be useful to prevent IUGR. The results indicate that both long- and short-term exposure to high-altitude causes disturbances in maternal ovarian steroidogenesis and negatively affects embryo-fetal growth already during the very early stages of gestation, with the consequences being even worsened in newcomers to high-altitude. The supply of antioxidant during this period only showed discrete effects for preventing IUGR. In conclusion, the present study gives a warning for clinicians about the risks for early-pregnant women when visiting high-altitude regions and suggests the need for further studies on the effects of the length of exposure and on the interaction of the exposure with the pregnancy stage.
Collapse
Affiliation(s)
- Victor H. Parraguez
- Faculty of Veterinary Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
- Faculty of Agricultural Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
- International Centre for Andean Studies, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Sandra Mamani
- Faculty of Agricultural Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Eileen Cofré
- Faculty of Veterinary Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Giorgio Castellaro
- Faculty of Agricultural Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Bessie Urquieta
- Faculty of Veterinary Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Mónica De los Reyes
- Faculty of Veterinary Sciences, University of Chile, Casilla 2, Correo 15, La Granja, Santiago, Chile
| | - Susana Astiz
- Comparative Physiology Lab, SGIT-INIA, Av. Puerta de Hierro s/n, 28040, Madrid, Spain
| | - Antonio Gonzalez-Bulnes
- Comparative Physiology Lab, SGIT-INIA, Av. Puerta de Hierro s/n, 28040, Madrid, Spain
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100, Sassari, Italy
| |
Collapse
|
47
|
Abstract
In spite of improving life expectancy over the course of the previous century, the health of the U.S. population is now worsening. Recent increasing rates of type 2 diabetes, obesity and uncontrolled high blood pressure predict a growing incidence of cardiovascular disease and shortened average lifespan. The daily >$1billion current price tag for cardiovascular disease in the United States is expected to double within the next decade or two. Other countries are seeing similar trends. Current popular explanations for these trends are inadequate. Rather, increasingly poor diets in young people and in women during pregnancy are a likely cause of declining health in the U.S. population through a process known as programming. The fetal cardiovascular system is sensitive to poor maternal nutritional conditions during the periconceptional period, in the womb and in early postnatal life. Developmental plasticity accommodates changes in organ systems that lead to endothelial dysfunction, small coronary arteries, stiffer vascular tree, fewer nephrons, fewer cardiomyocytes, coagulopathies and atherogenic blood lipid profiles in fetuses born at the extremes of birthweight. Of equal importance are epigenetic modifications to genes driving important growth regulatory processes. Changes in microRNA, DNA methylation patterns and histone structure have all been implicated in the cardiovascular disease vulnerabilities that cross-generations. Recent experiments offer hope that detrimental epigenetic changes can be prevented or reversed. The large number of studies that provide the foundational concepts for the developmental origins of disease can be traced to the brilliant discoveries of David J.P. Barker.
Collapse
|
48
|
Jonker SS, Giraud GD, Espinoza HM, Davis EN, Crossley DA. Effects of chronic hypoxia on cardiac function measured by pressure-volume catheter in fetal chickens. Am J Physiol Regul Integr Comp Physiol 2015; 308:R680-9. [PMID: 25652537 DOI: 10.1152/ajpregu.00484.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/02/2015] [Indexed: 11/22/2022]
Abstract
Hypoxia is a common component of many developmental insults and has been studied in early-stage chicken development. However, its impact on cardiac function and arterial-ventricular coupling in late-stage chickens is relatively unknown. To test the hypothesis that hypoxic incubation would reduce baseline cardiac function but protect the heart during acute hypoxia in late-stage chickens, white Leghorn eggs were incubated at 21% O2 or 15% O2. At 90% of incubation (19 days), hypoxic incubation caused growth restriction (-20%) and increased the LV-to-body ratio (+41%). Left ventricular (LV) pressure-volume loops were measured in anesthetized chickens in normoxia and acute hypoxia (10% O2). Hypoxic incubation lowered the maximal rate of pressure generation (ΔP/ΔtMax; -22%) and output (-57%), whereas increasing end-systolic elastance (ELV; +31%) and arterial elastance (EA; +122%) at similar heart rates to normoxic incubation. Both hypoxic incubation and acute hypoxia lengthened the half-time of relaxation (τ; +24%). Acute hypoxia reduced heart rate (-8%) and increased end-diastolic pressure (+35%). Hearts were collected for mRNA analysis. Hypoxic incubation was marked by decreased mRNA expression of sarco(endo)plasmic reticulum Ca(2+)-ATPase 2, Na(+)/Ca(2+) exchanger 1, phospholamban, and ryanodine receptor. In summary, hypoxic incubation reduces LV function in the late-stage chicken by slowing pressure generation and relaxation, which may be driven by altered intracellular excitation-contraction coupling. Cardiac efficiency is greatly reduced after hypoxic incubation. In both incubation groups acute hypoxia reduced diastolic function.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon;
| | - George D Giraud
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon; Veterans Affairs Portland Health Care System, Portland, Oregon; and
| | - Herbert M Espinoza
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Erica N Davis
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Dane A Crossley
- Department of Biological Sciences, University of North Texas, Denton, Texas
| |
Collapse
|
49
|
Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 2014; 224:164-75. [PMID: 25452175 DOI: 10.1016/j.cbi.2014.10.016] [Citation(s) in RCA: 908] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/13/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) initially considered as only damaging agents in living organisms further were found to play positive roles also. This paper describes ROS homeostasis, principles of their investigation and technical approaches to investigate ROS-related processes. Especial attention is paid to complications related to experimental documentation of these processes, their diversity, spatiotemporal distribution, relationships with physiological state of the organisms. Imbalance between ROS generation and elimination in favor of the first with certain consequences for cell physiology has been called "oxidative stress". Although almost 30years passed since the first definition of oxidative stress was introduced by Helmut Sies, to date we have no accepted classification of oxidative stress. In order to fill up this gape here classification of oxidative stress based on its intensity is proposed. Due to that oxidative stress may be classified as basal oxidative stress (BOS), low intensity oxidative stress (LOS), intermediate intensity oxidative stress (IOS), and high intensity oxidative stress (HOS). Another classification of potential interest may differentiate three categories such as mild oxidative stress (MOS), temperate oxidative stress (TOS), and finally severe (strong) oxidative stress (SOS). Perspective directions of investigations in the field include development of sophisticated classification of oxidative stresses, accurate identification of cellular ROS targets and their arranged responses to ROS influence, real in situ functions and operation of so-called "antioxidants", intracellular spatiotemporal distribution and effects of ROS, deciphering of molecular mechanisms responsible for cellular response to ROS attacks, and ROS involvement in realization of normal cellular functions in cellular homeostasis.
Collapse
Affiliation(s)
- Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Precarpathian National University named after Vassyl Stefanyk, 57 Shevchenko Str., Ivano-Frankivsk 76025, Ukraine.
| |
Collapse
|