1
|
Villarroel F, Ponce N, Gómez FA, Muñoz C, Ramírez E, Nualart F, Salinas P. Exposure to fine particulate matter 2.5 from wood combustion smoke causes vascular changes in placenta and reduce fetal size. Reprod Toxicol 2024; 127:108610. [PMID: 38750704 DOI: 10.1016/j.reprotox.2024.108610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
During gestation, maternal blood flow to the umbilical cord and placenta increases, facilitating efficient nutrient absorption, waste elimination, and effective gas exchange for the developing fetus. However, the effects of exposure to wood smoke during this period on these processes are unknown. We hypothesize that exposure to PM2.5, primarily sourced from wood combustion for home heating, affects placental vascular morphophysiology and fetal size. We used exposure chambers that received either filtered or unfiltered air. Female rats were exposed to PM2.5 during pre-gestational and/or gestational stages. Twenty-one days post-fertilization, placentas were collected via cesarean section. In these placentas, oxygen diffusion capacity was measured, and the expression of angiogenic factors was analyzed using qPCR and immunohistochemistry. In groups exposed to PM2.5 during pre-gestational and/or gestational stages, a decrease in fetal weight, crown-rump length, theoretical and specific diffusion capacity, and an increase in HIF-1α expression were observed. In groups exposed exclusively to PM2.5 during the pre-gestational stage, there was an increase in the expression of placental genes Flt-1, Kdr, and PIGF. Additionally, in the placental labyrinth region, the expression of angiogenic factors was elevated. Changes in angiogenesis and angiogenic factors reflect adaptations to hypoxia, impacting fetal growth and oxygen supply. In conclusion, this study demonstrates that exposure to PM2.5, emitted from wood smoke, in both pre-gestational and gestational stages, affects fetal development and placental health. This underscores the importance of addressing air pollution in areas with high levels of wood smoke, which poses a significant health risk to pregnant women and their fetuses.
Collapse
Affiliation(s)
- Francisca Villarroel
- Laboratory of Animal & Experimental Morphology, Institute of Biology, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile; MSc. Program in Biological Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Nikol Ponce
- PhD Program in Morphological Sciences, Universidad de La Frontera, Temuco, Chile; Center of Excellence in Surgical and Morphological Studies (CEMyQ), Universidad de La Frontera, Temuco, Chile
| | - Fernando A Gómez
- Laboratory of Genetics and Molecular Immunology, Institute of Biology, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cristián Muñoz
- Laboratory of Genetics and Molecular Immunology, Institute of Biology, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Eder Ramírez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepcion, Concepcion, Chile
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepcion, Concepcion, Chile; Center for Advanced Microscopy CMA BIO-BIO, Universidad de Concepcion, Concepcion, Chile
| | - Paulo Salinas
- Laboratory of Animal & Experimental Morphology, Institute of Biology, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
2
|
Lemieux P, Birot O. Altitude, Exercise, and Skeletal Muscle Angio-Adaptive Responses to Hypoxia: A Complex Story. Front Physiol 2021; 12:735557. [PMID: 34552509 PMCID: PMC8450406 DOI: 10.3389/fphys.2021.735557] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022] Open
Abstract
Hypoxia, defined as a reduced oxygen availability, can be observed in many tissues in response to various physiological and pathological conditions. As a hallmark of the altitude environment, ambient hypoxia results from a drop in the oxygen pressure in the atmosphere with elevation. A hypoxic stress can also occur at the cellular level when the oxygen supply through the local microcirculation cannot match the cells’ metabolic needs. This has been suggested in contracting skeletal myofibers during physical exercise. Regardless of its origin, ambient or exercise-induced, muscle hypoxia triggers complex angio-adaptive responses in the skeletal muscle tissue. These can result in the expression of a plethora of angio-adaptive molecules, ultimately leading to the growth, stabilization, or regression of muscle capillaries. This remarkable plasticity of the capillary network is referred to as angio-adaptation. It can alter the capillary-to-myofiber interface, which represent an important determinant of skeletal muscle function. These angio-adaptive molecules can also be released in the circulation as myokines to act on distant tissues. This review addresses the respective and combined potency of ambient hypoxia and exercise to generate a cellular hypoxic stress in skeletal muscle. The major skeletal muscle angio-adaptive responses to hypoxia so far described in this context will be discussed, including existing controversies in the field. Finally, this review will highlight the molecular complexity of the skeletal muscle angio-adaptive response to hypoxia and identify current gaps of knowledges in this field of exercise and environmental physiology.
Collapse
Affiliation(s)
- Pierre Lemieux
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Olivier Birot
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
3
|
Siragher E, Sferruzzi-Perri AN. Placental hypoxia: What have we learnt from small animal models? Placenta 2021; 113:29-47. [PMID: 34074553 DOI: 10.1016/j.placenta.2021.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 12/31/2022]
Abstract
Intrauterine hypoxia is a feature of pregnancy complications, both at high altitude and sea level. To understand the placental response to reduced oxygen availability, small animal models of maternal inhalation hypoxia (MIH) or reduced uterine perfusion pressure (RUPP) may be utilised. The aim of this review was to compare the findings of those studies to identify the role of oxygen availability in adapting placental structural and functional phenotypes in relation to fetal outcome. It also sought to explore the evidence for the involvement of particular genes and protein signalling pathways in the placenta in mediating hypoxia driven alterations. The data available demonstrate that both MIH and RUPP can induce placental hypoxia, which affects placental structure and vascularity, as well as glucose, amino acid, calcium and possibly lipid transport capacity. In addition, changes have been observed in HIF, VEGF, insulin/IGF2, AMPK, mTOR, PI3K and PPARγ signalling, which may be key in linking together observed phenotypes under conditions of placental hypoxia. Many different manipulations have been examined, with varied outcomes depending on the intensity, timing and duration of the insult. Some manipulations have detrimental effects on placental phenotype, viability and fetal growth, whereas in others, the placenta appears to adapt to uphold fetal growth despite the challenge of low oxygen. Together these data suggest a complex response of the placenta to reduced oxygen availability, which links to changes in fetal outcomes. However, further work is required to explore the role of fetal sex, altered maternal physiology and placental molecular mechanisms to fully understand placental responses to hypoxia and their relevance for pregnancy outcome.
Collapse
Affiliation(s)
- Emma Siragher
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
4
|
Kobayashi T, Ueda S, Takagi M, Kihara M, Suzuki Y. Pathophysiological roles of ADMA-mediated endothelial injury in hypertensive disorders of pregnancy. HYPERTENSION RESEARCH IN PREGNANCY 2020. [DOI: 10.14390/jsshp.hrp2019-013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | - Seiji Ueda
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Miyuki Takagi
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Masao Kihara
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine
| |
Collapse
|
5
|
Malaria in Pregnancy and Adverse Birth Outcomes: New Mechanisms and Therapeutic Opportunities. Trends Parasitol 2019; 36:127-137. [PMID: 31864896 DOI: 10.1016/j.pt.2019.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022]
Abstract
Malaria infection during pregnancy is associated with adverse birth outcomes but underlying mechanisms are poorly understood. Here, we discuss the impact of malaria in pregnancy on three pathways that are important regulators of healthy pregnancy outcomes: L-arginine-nitric oxide biogenesis, complement activation, and the heme axis. These pathways are not mutually exclusive, and they collectively create a proinflammatory, antiangiogenic milieu at the maternal-fetal interface that interferes with placental function and development. We hypothesize that targeting these host-response pathways would mitigate the burden of adverse birth outcomes attributable to malaria in pregnancy.
Collapse
|
6
|
McDonald CR, Cahill LS, Gamble JL, Elphinstone R, Gazdzinski LM, Zhong KJY, Philson AC, Madanitsa M, Kalilani-Phiri L, Mwapasa V, Ter Kuile FO, Sled JG, Conroy AL, Kain KC. Malaria in pregnancy alters l-arginine bioavailability and placental vascular development. Sci Transl Med 2019. [PMID: 29514999 PMCID: PMC6510298 DOI: 10.1126/scitranslmed.aan6007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reducing adverse birth outcomes due to malaria in pregnancy (MIP) is a global health priority. However, there are few safe and effective interventions. L-arginine is an essential amino acid in pregnancy and an immediate precursor in the biosynthesis of nitric oxide (NO), but there are limited data on the impact of MIP on NO biogenesis. We hypothesized that hypoarginemia contributes to the pathophysiology of MIP and that L-arginine supplementation would improve birth outcomes. In a prospective study of pregnant Malawian women, we show that MIP was associated with lower concentrations of L- arginine and higher concentrations of endogenous inhibitors of NO biosynthesis, asymmetric and symmetric dimethylarginine, which were associated with adverse birth outcomes. In a model of experimental MIP, L-arginine supplementation in dams improved birth outcomes (decreased stillbirth and increased birth weight) compared with controls. The mechanism of action was via normalized angiogenic pathways and enhanced placental vascular development, as visualized by placental microcomputerized tomography imaging. These data define a role for dysregulation of NO biosynthetic pathways in the pathogenesis of MIP and support the evaluation of interventions to enhance L-arginine bioavailability as strategies to improve birth outcomes.
Collapse
Affiliation(s)
- Chloe R McDonald
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Joel L Gamble
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Robyn Elphinstone
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Lisa M Gazdzinski
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Kathleen J Y Zhong
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Adrienne C Philson
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02138, USA
| | | | | | - Victor Mwapasa
- College of Medicine, University of Malawi, P.O. Box 280, Blantyre, Malawi
| | | | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Andrea L Conroy
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Kevin C Kain
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada. .,Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
7
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
8
|
Kim SM, Yun HG, Kim RY, Chung YH, Cheon JY, Wie JH, Kwon JY, Ko HS, Kim YH, Han EH, Park JH, Kim HJ, Kim MS, Shin JC, Park IY. Maternal serum placental growth factor combined with second trimester aneuploidy screening to predict small-for-gestation neonates without preeclampsia. Taiwan J Obstet Gynecol 2017; 56:801-805. [DOI: 10.1016/j.tjog.2017.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2017] [Indexed: 11/25/2022] Open
|
9
|
Potential New Non-Invasive Therapy Using Artificial Oxygen Carriers for Pre-Eclampsia. J Funct Biomater 2017; 8:jfb8030032. [PMID: 28758949 PMCID: PMC5618283 DOI: 10.3390/jfb8030032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/16/2017] [Accepted: 07/18/2017] [Indexed: 01/10/2023] Open
Abstract
The molecular mechanisms of pre-eclampsia are being increasingly clarified in animals and humans. With the uncovering of these mechanisms, preventive therapy strategies using chronic infusion of adrenomedullin, vascular endothelial growth factor-121 (VEGF-121), losartan, and sildenafil have been proposed to block narrow spiral artery formation in the placenta by suppressing related possible factors for pre-eclampsia. However, although such preventive treatments have been partly successful, they have failed in ameliorating fetal growth restriction and carry the risk of possible side-effects of drugs on pregnant mothers. In this study, we attempted to develop a new symptomatic treatment for pre-eclampsia by directly rescuing placental ischemia with artificial oxygen carriers (hemoglobin vesicles: HbV) since previous data indicate that placental ischemia/hypoxia may alone be sufficient to lead to pre-eclampsia through up-regulation of sFlt-1, one of the main candidate molecules for the cause of pre-eclampsia. Using a rat model, the present study demonstrated that a simple treatment using hemoglobin vesicles for placental ischemia rescues placental and fetal hypoxia, leading to appropriate fetal growth. The present study is the first to demonstrate hemoglobin vesicles successfully decreasing maternal plasma levels of sFlt-1 and ameliorating fetal growth restriction in the pre-eclampsia rat model (p < 0.05, one-way ANOVA). In future, chronic infusion of hemoglobin vesicles could be a potential effective and noninvasive therapy for delaying or even alleviating the need for Caesarean sections in pre-eclampsia.
Collapse
|
10
|
Chauhan M, Balakrishnan M, Vidaeff A, Yallampalli U, Lugo F, Fox K, Belfort M, Yallampalli C. Adrenomedullin2 (ADM2)/Intermedin (IMD): A Potential Role in the Pathophysiology of Preeclampsia. J Clin Endocrinol Metab 2016; 101:4478-4488. [PMID: 27583470 PMCID: PMC5095259 DOI: 10.1210/jc.2016-1333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT It is not known whether decreases in trophoblast invasion promoting the peptide, adrenomedullin2 (ADM2) system is associated with preeclampsia (PreE). OBJECTIVE The objective of the study was to assess the changes in ADM2 levels in plasma, placenta, and amniotic fluid (AF) and its receptor components in placenta from PreE pregnancy compared with the age-matched normal and study the effect of ADM2 on the synthesis of nitric oxide (NO), endothelial nitric oxide synthase (eNOS), and matrix-metalloproteinase (MMP)-2 and MMP-9 in trophoblast cells. RESULTS PreE is associated with a decreased expression of ADM2 in plasma and placenta (P < .05); ADM2 interacts with a seven-transmembrane G protein-coupled receptor, calcitonin receptor-like receptor (CRLR) in HTR-8/SVneo cells; placental expression of ADM2/CRLR complex is lower in PreE; mRNA for CRLR and receptor activity-modifying protein-3 are lower, whereas receptor activity-modifying protein-2 is higher in the PreE placenta (P < .05); ADM2 levels in the second trimester are lower in the AF from pregnant women who develop PreE later in gestation (P < .05); ADM2 is localized to the epithelium of the amnion and the ectoderm and mesoderm of the chorion in term fetal membranes; ADM2 increases NO production, eNOS, and MMP2/9-immunoreactivity, whereas ADM2 knockdown inhibits the expression of eNOS and MMP2/9 mRNA and S-nitrosylation in HTR-8/SVneo cells; and ADM2-induced increases in MMP2/9 activity is inhibited by L-nitro-arginine methyl ester in HTR-8SV/neo cells. CONCLUSION Decreases in the ADM2 system in PreE at term, in AF from pregnant women during the second trimester who develop PreE later in gestation, and ADM2-induced increases in the NO and MMP-2/9 levels in trophoblast cells suggest a potential role for ADM2 via the NO-MMP system in the pathophysiology of PreE.
Collapse
Affiliation(s)
- Madhu Chauhan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Meena Balakrishnan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Alex Vidaeff
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Uma Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Fernando Lugo
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Karin Fox
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Michael Belfort
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| | - Chandra Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
11
|
Artificial oxygen carriers rescue placental hypoxia and improve fetal development in the rat pre-eclampsia model. Sci Rep 2015; 5:15271. [PMID: 26471339 PMCID: PMC4608007 DOI: 10.1038/srep15271] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/17/2015] [Indexed: 01/17/2023] Open
Abstract
Pre-eclampsia affects approximately 5% of all pregnant women and remains a major cause of maternal and fetal morbidity and mortality. The hypertension associated with pre-eclampsia develops during pregnancy and remits after delivery, suggesting that the placenta is the most likely origin of this disease. The pathophysiology involves insufficient trophoblast invasion, resulting in incomplete narrow placental spiral artery remodeling. Placental insufficiency, which limits the maternal-fetal exchange of gas and nutrients, leads to fetal intrauterine growth restriction. In this study, in our attempt to develop a new therapy for pre-eclampsia, we directly rescued placental and fetal hypoxia with nano-scale size artificial oxygen carriers (hemoglobin vesicles). The present study is the first to demonstrate that artificial oxygen carriers successfully treat placental hypoxia, decrease maternal plasma levels of anti-angiogenic proteins and ameliorate fetal growth restriction in the pre-eclampsia rat model.
Collapse
|
12
|
Harrison JF, Shingleton AW, Callier V. Stunted by Developing in Hypoxia: Linking Comparative and Model Organism Studies. Physiol Biochem Zool 2015; 88:455-70. [PMID: 26658244 DOI: 10.1086/682216] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Animals develop in atmospheric hypoxia in a wide range of habitats, and tissues may experience O2 limitation of ATP production during postembryonic development if O2 supply structures do not keep pace with growing O2 demand during ontogeny. Most animal species are stunted by postembryonic development in hypoxia, showing reduced growth rates and size in moderate hypoxia (5-15 kPa Po2). In mammals, the critical Po2 that limits resting metabolic rate also falls in this same moderate hypoxic range, so stunted growth may simply be due to hypoxic limits on ATP production. However, in most invertebrates and at least some lower vertebrates, hypoxic stunting occurs at Po2 values well above those that limit resting metabolism. Studies with diverse model organisms have identified multiple homologous O2-sensing signaling pathways that can inhibit feeding and growth during moderate hypoxia. Together, these comparative and model organism-based studies suggest that hypoxic stunting of growth and size can occur as programmed inhibition of growth, often by inhibition of insulin stimulation of growth processes. Furthermore, there is increasing evidence that these same O2 signaling pathways can be utilized during normal animal development to ensure matching of O2 supply and demand structures and in mediation of variation in animal performance.
Collapse
Affiliation(s)
- Jon F Harrison
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287; 2Department of Biology, Lake Forest College, Lake Forest, Illinois 60045
| | | | | |
Collapse
|
13
|
Jang EA, Longo LD, Goyal R. Antenatal maternal hypoxia: criterion for fetal growth restriction in rodents. Front Physiol 2015; 6:176. [PMID: 26106333 PMCID: PMC4458570 DOI: 10.3389/fphys.2015.00176] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/25/2015] [Indexed: 12/24/2022] Open
Abstract
Rodents are a useful model for life science research. Accumulating evidence suggests that the offspring of mice and rats suffer from similar disorders as humans when exposed to hypoxia during pregnancy. Importantly, with antenatal hypoxic exposure, human neonates demonstrate low birth weight or growth restriction. Similarly, with antenatal hypoxic exposure rodents also demonstrate the fetal growth restriction (FGR). Surprisingly, there is no consensus on the minimum duration or degree of hypoxic exposure required to cause FGR in rodents. Thus, we have reviewed the available literature in an attempt to answer these questions. Based on studies in rats, birth weight reduction of 31% corresponded to 10th percentile reduction in birth weight curve. With the similar criterion (10th percentile), in mice 3 days or more and in rats 7 days or more of 14% or lower hypoxia administration was required to produce statistically significant FGR.
Collapse
Affiliation(s)
- Eeun Amy Jang
- Department of Basic Sciences, Center for Perinatal Biology, School of Medicine, Loma Linda University Loma Linda, CA, USA
| | - Lawrence D Longo
- Department of Basic Sciences, Center for Perinatal Biology, School of Medicine, Loma Linda University Loma Linda, CA, USA ; Epigenuity LLC Loma Linda, CA, USA
| | - Ravi Goyal
- Department of Basic Sciences, Center for Perinatal Biology, School of Medicine, Loma Linda University Loma Linda, CA, USA ; Epigenuity LLC Loma Linda, CA, USA
| |
Collapse
|
14
|
Nayeri UA, Buhimschi IA, Laky CA, Cross SN, Duzyj CM, Ramma W, Sibai BM, Funai EF, Ahmed A, Buhimschi CS. Antenatal Corticosteroids Impact the Inflammatory Rather Than the Antiangiogenic Profile of Women With Preeclampsia. Hypertension 2014; 63:1285-92. [DOI: 10.1161/hypertensionaha.114.03173] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Unzila A. Nayeri
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Irina A. Buhimschi
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Christine A. Laky
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Sarah N. Cross
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Christina M. Duzyj
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Wenda Ramma
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Baha M. Sibai
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Edmund F. Funai
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Asif Ahmed
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| | - Catalin S. Buhimschi
- From the Department of Obstetrics and Gynecology, Upstate Medical University, Syracuse, NY (U.A.N.); Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH (I.A.B., C.S.B.); Departments of Pediatrics (I.A.B., C.S.B.) and Obstetrics and Gynecology (I.A.B., E.F.F., C.S.B.), The Ohio State University College of Medicine, Columbus; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT (C.A.L., S.N.C., C.M.D., W.R.)
| |
Collapse
|
15
|
Cuffe JSM, Walton SL, Singh RR, Spiers JG, Bielefeldt-Ohmann H, Wilkinson L, Little MH, Moritz KM. Mid- to late term hypoxia in the mouse alters placental morphology, glucocorticoid regulatory pathways and nutrient transporters in a sex-specific manner. J Physiol 2014; 592:3127-41. [PMID: 24801305 DOI: 10.1113/jphysiol.2014.272856] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Maternal hypoxia is a common perturbation that can disrupt placental and thus fetal development, contributing to neonatal impairments. Recently, evidence has suggested that physiological outcomes are dependent upon the sex of the fetus, with males more susceptible to hypoxic insults than females. This study investigated the effects of maternal hypoxia during mid- to late gestation on fetal growth and placental development and determined if responses were sex specific. CD1 mice were housed under 21% or 12% oxygen from embryonic day (E) 14.5 until tissue collection at E18.5. Fetuses and placentas were weighed before collection for gene and protein expression and morphological analysis. Hypoxia reduced fetal weight in both sexes at E18.5 by 7% but did not affect placental weight. Hypoxia reduced placental mRNA levels of the mineralocorticoid and glucocorticoid receptors and reduced the gene and protein expression of the glucocorticoid metabolizing enzyme HSD11B2. However, placentas of female fetuses responded differently to maternal hypoxia than did placentas of male fetuses. Notably, morphology was significantly altered in placentas from hypoxic female fetuses, with a reduction in placental labyrinth blood spaces. In addition mRNA expression of Glut1, Igf2 and Igf1r were reduced in placentas of female fetuses only. In summary, maternal hypoxia altered placental formation in a sex specific manner through mechanisms involving placental vascular development, growth factor and nutrient transporter expression and placental glucocorticoid signalling. This study provides insight into how sex differences in offspring disease development may be due to sex specific placental adaptations to maternal insults.
Collapse
Affiliation(s)
- J S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - S L Walton
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - R R Singh
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - J G Spiers
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - H Bielefeldt-Ohmann
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - L Wilkinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - M H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
16
|
Tarrade A, Lecarpentier E, Gil S, Morel O, Zahr N, Dahirel M, Tsatsaris V, Chavatte-Palmer P. Analysis of placental vascularization in a pharmacological rabbit model of IUGR induced by l-NAME, a nitric oxide synthase inhibitor. Placenta 2014; 35:254-9. [DOI: 10.1016/j.placenta.2014.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 11/25/2022]
|
17
|
Weissgerber TL, McConico A, Knudsen BE, Butters KA, Hayman SR, White WM, Milic N, Miller VM, Garovic VD. Methodological differences account for inconsistencies in reported free VEGF concentrations in pregnant rats. Am J Physiol Regul Integr Comp Physiol 2014; 306:R796-803. [PMID: 24622973 DOI: 10.1152/ajpregu.00544.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Free vascular endothelial growth factor (VEGF) is undetectable in plasma during human pregnancy. However, studies examining pregnant rats have reported both low (8-29 pg/ml) and high (527-1,030 pg/ml) free VEGF. These discrepancies cast uncertainty over the use of rat models to study angiogenic factors in pregnancy and preeclampsia. This study investigates methodological factors that may explain these discrepancies. Plasma VEGF in nonpregnant, day 7 pregnant, and day 19 pregnant rats was measured using rat and mouse ELISAs (R&D Systems). The rat ELISA detected VEGF in plasma from nonpregnant rats but not in plasma from day 19 pregnant rats. The mouse ELISA detected higher VEGF concentrations than the rat ELISA in every sample tested. This discrepancy was greater in day 19 pregnant rats (median: 2,273 vs. 0 pg/ml) than in nonpregnant (97 vs. 20 pg/ml) and day 7 pregnant (66 vs. 2 pg/ml) rats. Recovery of recombinant rat VEGF (rrVEGF) spiked into plasma from nonpregnant and day 7 pregnant rats was high for the rat ELISA (82-105%) but low for the mouse ELISA (17-22%). The rat ELISA did not recover rrVEGF in plasma from day 19 pregnant rats, suggesting that this ELISA measures free VEGF. The use of the rat versus mouse ELISA likely explains the differences in reported VEGF concentrations in pregnant rats. While the rat ELISA appears to measure free VEGF, plasma concentrations in nonpregnant and pregnant rats are below the assay sensitivity limit. As most previous studies of pregnant rats used the mouse VEGF ELISA, these data should be interpreted cautiously.
Collapse
Affiliation(s)
| | | | - Bruce E Knudsen
- Department of Experimental Pathology and Lab Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kim A Butters
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Wendy M White
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Natasa Milic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; Department of Biostatistics, Belgrade Medical School, Belgrade, Serbia; and
| | - Virginia M Miller
- Departments of Surgery, Physiology and Biomedical Engineering Mayo Clinic, Rochester, Minnesota
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Zamudio S, Borges M, Echalar L, Kovalenko O, Vargas E, Torricos T, Khan AA, Alvarez M, Illsley NP. Maternal and fetoplacental hypoxia do not alter circulating angiogenic growth effectors during human pregnancy. Biol Reprod 2014; 90:42. [PMID: 24352559 DOI: 10.1095/biolreprod.113.115592] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
One causal model of preeclampsia (PE) postulates that placental hypoxia alters the production of angiogenic growth effectors (AGEs), causing an imbalance leading to maternal endothelial cell dysfunction. We tested this model using the natural experiment of high-altitude (HA) residence. We hypothesized that in HA pregnancies 1) circulating soluble fms-like tyrosine kinase 1 (sFlt-1) is increased and placental growth factor (PlGF) decreased, and 2) AGE concentrations correlate with measures of hypoxia. A cross-sectional study of healthy pregnancies at low altitude (LA) (400 m) versus HA (3600 m) compared normal (n = 80 at HA, n = 90 at LA) and PE pregnancies (n = 20 PE at HA, n = 19 PE at LA). Blood was collected using standard serum separation and, in parallel, by a method designed to inhibit platelet activation. AGEs were measured by enzyme-linked immunosorbent assays. AGEs did not differ between altitudes in normal or PE pregnancies. AGE concentrations were unrelated to measures of maternal or fetal hypoxia. PlGF was lower and sFlt-1 higher in PE, but overlapped considerably with the range observed in normal samples. PlGF correlated with placental mass in both normal and PE pregnancies. The contribution of peripheral cells to the values measured for AGEs was similar at LA and HA, but was greater in PE than in normotensive women. Hypoxia, across a wide physiological range in pregnancy, does not alter levels of circulating AGEs in otherwise normal pregnancies. Peripheral cell release of AGEs with the hemostasis characteristic of standard blood collection is highly variable and contributes to a doubling of the amount of sFlt-1 measured in PE as compared to normal pregnancies.
Collapse
Affiliation(s)
- Stacy Zamudio
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery, Hackensack University Medical Center, Hackensack, New Jersey
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Review: Potential druggable targets for the treatment of early onset preeclampsia. Pregnancy Hypertens 2013; 3:203-10. [DOI: 10.1016/j.preghy.2013.04.120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 04/29/2013] [Indexed: 12/29/2022]
|
20
|
Serum levels of asymmetric dimethylarginine, vascular endothelial growth factor, and nitric oxide metabolite levels in preeclampsia patients. ISRN OBSTETRICS AND GYNECOLOGY 2013; 2013:104213. [PMID: 24109515 PMCID: PMC3786521 DOI: 10.1155/2013/104213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/24/2013] [Indexed: 01/24/2023]
Abstract
Background. Hypertensive disorder generally complicates 5-10 percent of all pregnancies. Angiogenic growth factors may be helpful for the diagnosis and prediction of preeclampsia. Therefore, in this study we attempted to determine the serum levels of asymmetric dimethylarginine (ADMA), vascular endothelial growth factor (VEGF), and nitric oxide (NO) metabolite (nitrite) in preeclampsia patients and compared the levels with those obtained from normal pregnant women. Methods. Ninety pregnant women (19-33 years old) in two groups of preeclampsia and normal were considered during 2012. The levels of ADMA, VEGF, and nitrite were measured in maternal serum samples using ELISA kits. Results. Significant increase of VEGF and nitrite levels was observed in preeclampsia patients when compared with other groups (P < 0.05). The serum level of ADMA demonstrated a similar increased trend in preeclampsia patients; however, the increase was not statistically significant (P = 0.08). Conclusion. The findings reveal that the elevation of serum levels of VEGF and nitrite and possibly ADMA may be involved in the pathogenesis of preeclampsia.
Collapse
|
21
|
Kulandavelu S, Whiteley KJ, Qu D, Mu J, Bainbridge SA, Adamson SL. Endothelial nitric oxide synthase deficiency reduces uterine blood flow, spiral artery elongation, and placental oxygenation in pregnant mice. Hypertension 2012; 60:231-8. [PMID: 22615111 DOI: 10.1161/hypertensionaha.111.187559] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Preeclampsia is associated with impaired uteroplacental adaptations during pregnancy and abnormalities in the endothelial NO synthase (eNOS)-NO pathway, but whether eNOS deficiency plays a causal role is unknown. Thus, the objective of the current study was to determine the role of eNOS in the mother and/or conceptus in uteroplacental changes during pregnancy using eNOS knockout mice. We quantified uterine artery blood flow using microultrasound, visualized the uteroplacental vasculature using vascular corrosion casts, and used pimonidazole and hypoxia-inducible factor 1α immunohistochemistry as markers of hypoxia in the placentas of eNOS knockout mice versus the background strain, C57Bl/6J (wild type). We found that increases in uteroplacental blood flow, uterine artery diameter, and spiral artery length were reduced, and markers of placental hypoxia in the junctional zone were elevated in late gestation in eNOS knockout mice. Both maternal and conceptus genotypes contributed to changes in uterine artery diameter and flow. Despite placental hypoxia, placental soluble fms-like tyrosine kinase 1 and tumor necrosis factor-α mRNA, and in maternal plasma, soluble fms-like tyrosine kinase 1 were not elevated in eNOS knockout mice. Thus, our results show that both eNOS in the mother and the conceptus contribute to uteroplacental vascular changes and increased uterine arterial blood flow in normal pregnancy.
Collapse
|
22
|
Severely decreased activity of placental dimethylarginine dimethylaminohydrolase in pre-eclampsia. Eur J Obstet Gynecol Reprod Biol 2012; 161:152-6. [PMID: 22285683 DOI: 10.1016/j.ejogrb.2011.12.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 11/28/2011] [Accepted: 12/21/2011] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Asymmetric dimethylarginine (ADMA) is a key regulator of nitric oxide production. Elevations of ADMA have previously been associated with endothelial dysfunction in pre-eclamptic women. ADMA is degraded mainly by dimethylarginine dimethylaminohydrolase (DDAH), which is also expressed in placental tissue. Therefore, we measured placental DDAH expression and activity in pre-eclampsia and normal pregnancies in order to determine whether impairment of this enzyme in the pre-eclamptic placenta could contribute to elevations of ADMA levels in these women. STUDY DESIGN ADMA plasma levels were measured by LC-MS/MS in 18 pre-eclamptic patients and 28 controls. Placental DDAH activity was determined by measuring the degradation of [(2)H(6)]-labeled ADMA in tissue homogenates from placental biopsies in 15 women with pre-eclampsia and 16 controls. Placental mRNA expression of DDAH 1, DDAH 2, endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS) and protein-arginine methyltransferase 1 (PRMT1) was determined in tissue biopsies by RT-PCR. RESULTS Placental DDAH activity was almost undetectable in pre-eclampsia, and it was significantly higher in controls. ADMA plasma levels were higher in pre-eclampsia as compared to normal pregnancies (0.51±0.15μmol/l vs. 0.42±0.07μmol/l; p=0.005), and the difference between maternal and fetal ADMA levels (feto-maternal ADMA gradient) was lower in pre-eclampsia (0.63±0.20μmol/l vs. 0.80±0.18μmol/l; p=0.02). Furthermore, mRNA expression levels of DDAH 2 were significantly lower in pre-eclamptic women (p=0.04), while PRMT1 expression levels were the same. In pre-eclampsia, we found only weak correlations between maternal ADMA levels and DDAH 1 (r=-0.41; p=0.22) and DDAH 2 expressions (r=-0.45; p=0.17) but a slightly stronger correlation between DDAH 2 expression and feto-maternal ADMA gradient (r=0.60; p=0.07). CONCLUSION Decreased DDAH activity in the pre-eclamptic placenta might contribute to elevated ADMA levels in these patients.
Collapse
|
23
|
Pang LH, Li MJ, Li MQ, Yang DM, Shi L. Vascular endothelial growth factor (VEGF) and the VEGF soluble receptor-1 (sFlt-1) in chorionic villus tissue from Chinese women with early recurrent spontaneous abortion. J Int Med Res 2011; 39:830-7. [PMID: 21819715 DOI: 10.1177/147323001103900316] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This case-control study explored the relationship between early recurrent spontaneous abortion (RSA) and the expression of two genes: VEGFA, the gene encoding vascular endothelial growth factor (VEGF); and fms-related tyrosine kinase 1 (FLT1), the gene encoding the soluble VEGF receptor-1 (sFlt-1). Women experiencing RSA or undergoing induced abortions in the early stage of normal pregnancy were recruited to the study (n = 30 per group). There were no significant between-group differences in maternal age or duration of pregnancy. The levels of VEGF and sFlt-1 mRNA in chorionic villus tissue samples were examined by quanti tative reverse transcription-polymerase chain reaction. Levels of sFlt-1 and VEGF mRNA in the chorionic villus tissue of women with RSA were significantly higher than levels in the control group. This study demonstrated that there is a relationship between early RSA and VEGF and sFlt-1 levels, suggesting that over-expression of the FLT1 and VEGFA genes may be associated with the pathogenesis of RSA.
Collapse
Affiliation(s)
- L-H Pang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | | | | | | |
Collapse
|
24
|
Groesch KA, Torry RJ, Wilber AC, Abrams R, Bieniarz A, Guilbert LJ, Torry DS. Nitric oxide generation affects pro- and anti-angiogenic growth factor expression in primary human trophoblast. Placenta 2011; 32:926-31. [PMID: 21963217 DOI: 10.1016/j.placenta.2011.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/17/2011] [Accepted: 08/18/2011] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Preeclampsia is associated with reduced trophoblast placenta growth factor (PGF) expression, elevated soluble fms-like tyrosine kinase-1 (sFlt-1) and decreased bioactivity of nitric oxide (NO). Elevated sFlt-1 reduces bio-availability of PGF and vascular endothelial growth factor (VEGF) leading to maternal endothelial dysfunction. Although NO can regulate gene expression, its ability to regulate trophoblast expression of angiogenic growth factors is not known. STUDY DESIGN Human primary term trophoblast and JEG-3 choriocarcinoma cells were cultured under 21%O(2) or 1%O(2) conditions in the presence or absence of NO donor (SNP) or inhibitor (L-NAME). Effects on PGF, VEGF and Flt-1 isoform mRNA expression were determined by quantitative real-time PCR. Changes in expression of soluble protein isoforms of FLT-1 was monitored by ELISA. RESULTS Hypoxia decreased PGF mRNA but increased VEGF, sFlt-1 and Flt-1 mRNA expression in trophoblast. Generation of NO in trophoblast under 1%O(2) culture conditions significantly reversed sFlt-1 mRNA and protein expression, independent of mFlt-1. Conversely NO generation in hypoxic trophoblast increased VEGF and PGF mRNA expression. CONCLUSIONS NO production in primary human trophoblast cultures had divergent effects on pro-angiogenic (PGF, VEGF) versus anti-angiogenic (sFlt-1) mRNA expression, resulting in an enhanced pro-angiogenic gene expression environment in vitro.
Collapse
Affiliation(s)
- K A Groesch
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Dávila RD, Julian CG, Wilson MJ, Browne VA, Rodriguez C, Bigham AW, Shriver MD, Vargas E, Moore LG. Do anti-angiogenic or angiogenic factors contribute to the protection of birth weight at high altitude afforded by Andean ancestry? Reprod Sci 2010; 17:861-70. [PMID: 20601535 DOI: 10.1177/1933719110372418] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE This prospective study was designed to determine whether variation in angiogenic (placental growth factor [PlGF]) and/or anti-angiogenic (soluble fms-like tyrosine kinase [sFlt-1]) factors contribute to the protective effect of highland ancestry (Andean) from altitude-associated reductions in fetal growth. STUDY DESIGN Plasma sFlt-1 and PlGF levels, uterine artery (UA) blood flow, and fetal biometry were determined in low-altitude (400 m; Andean n = 27, European n = 28) and high-altitude (3600 m; Andean n = 51, European n = 44) residents during pregnancy (20 and 36 weeks) and 4 months postpartum. RESULTS High-altitude decreased sFlt-1 levels in both groups, Andeans had lower sFlt-1, comparable PlGF, lower sFlt-1/PlGF ratios, and higher UA blood flow throughout pregnancy relative to Europeans. Altitude decreased birth weight in Europeans but not Andeans. In high-altitude Europeans sFlt-1/PlGF and sFlt-1 levels were negatively associated with UA diameter and birth weight, respectively. CONCLUSIONS Lower sFlt-1 and sFlt-1/PLGF ratio may contribute to or result from variations in maternal vascular adaptation to pregnancy between Andean and Europeans at high altitude. Subsequently, these effects could potentially influence ancestry-associated differences in birth weight.
Collapse
Affiliation(s)
- R Daniela Dávila
- Altitude Research Center, University of Colorado-Denver, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gobble RM, Groesch KA, Chang M, Torry RJ, Torry DS. Differential regulation of human PlGF gene expression in trophoblast and nontrophoblast cells by oxygen tension. Placenta 2009; 30:869-75. [PMID: 19712973 DOI: 10.1016/j.placenta.2009.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 08/05/2009] [Accepted: 08/06/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To determine the mechanism for differential effects of low oxygen tension on human PlGF gene transcription in trophoblast and nontrophoblast cells. STUDY DESIGN Human PlGF reporter clones and real-time RT-PCR were used to compare the effects of hypoxia on gene transcription in human trophoblast and nontrophoblast cell lines. Overexpression of HIF-1alpha, inhibition of HIF-1 function and biochemical assessments of HIF-1 co-factor interactions were used to characterize hypoxia response mechanisms regulating PlGF transcription. RESULTS PlGF transcription is specifically inhibited by low oxygen tension in trophoblast but is induced in some nontrophoblast cells. Overexpression of HIF-1alpha in normoxic cells or inhibition of HIF-1 function in hypoxic cells did not significantly alter transcription patterns of the PlGF gene in either cell type. CONCLUSIONS These results suggest that transcriptional repression of PlGF gene expression occurs in human trophoblast exposed to low oxygen tension but that PlGF transcription is stimulated in certain hypoxic nontrophoblast cells. However, regulation of PlGF transcription is not mediated by functional HIF-1 activity in either cell type.
Collapse
Affiliation(s)
- R M Gobble
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA
| | | | | | | | | |
Collapse
|
27
|
Guller S. Role of the syncytium in placenta-mediated complications of preeclampsia. Thromb Res 2009; 124:389-92. [PMID: 19535132 DOI: 10.1016/j.thromres.2009.05.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 05/22/2009] [Accepted: 05/22/2009] [Indexed: 01/02/2023]
Abstract
The syncytiotrophoblast (SCT) is the outer layer of placenta which is in direct contact with maternal blood. As such it is uniquely positioned to alter maternal hemostasis and endothelial function. The syncytium is known to release anti-angiogenic factors including fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), as well as the anti-fibrinolytic factor plasminogen activator inhibitor-1 (PAI-1). Its release of microparticles has also been suggested to play a role in regulating maternal endothelial and immune cell function. It is of note that syncytial release of the above-mentioned factors increases in preeclampsia, a major cause of maternal mortality and morbidity. In preeclampsia, hypoxia and reperfusion injury in the placenta is associated with activation of the maternal endothelium. In this review, I describe the interaction of syncytial factors with hypoxia, reactive oxygen species, and apoptosis in the pathophysiology of preeclampsia and intrauterine growth restriction. In addition, I detail the potential protective actions of placental ceruloplasmin in preeclampsia, recently described by our group to be a sensitive marker of syncytial hypoxia.
Collapse
Affiliation(s)
- Seth Guller
- Department of Obstetrics/Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520-8063, USA
| |
Collapse
|
28
|
Sfakianaki AK, Buhimschi IA, Ravishankar V, Bahtiyar MO, Dulay AT, Buhimschi CS. Relationships of maternal serum levels of vascular endothelial growth factor and tensile strength properties of the cervix in a rat model of chronic hypoxia. Am J Obstet Gynecol 2008; 198:223.e1-7. [PMID: 18226631 DOI: 10.1016/j.ajog.2007.09.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 07/23/2007] [Accepted: 09/27/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVE It has been shown that hypoxia leads to alterations in maternal serum levels of vascular endothelial growth factor (VEGF). In this study, we sought to test the hypothesis that chronic hypoxia increases maternal serum levels of VEGF, which in turn cause measurable changes in the viscoelastic properties of the rat uterine cervix. STUDY DESIGN Timed-pregnant adult Sprague-Dawley rats were exposed to hypoxia beginning on day 17 of gestation (term = day 22). The following groups of animals were studied: (1) nonpregnant controls (NP, n = 6); (2) normoxia 21% fraction of inspired oxygen (FiO2) (NMX, n = 6); and (3) severe hypoxia 10% FiO2 (HPX; n = 5). A hypoxic chamber was used to assure consistent hypoxic environment. Animals were killed on day 21 of gestation (before labor). Maternal blood was collected immediately following anesthesia and prior to euthanasia. Free serum levels of VEGF were measured by highly specific immunoassays. Tensile strength properties of the cervix were assessed using a stretching regimen designed to mimic labor. Physical parameters measured were: indicators of viscoelasticity (slope; measure of stiffness), plasticity (yield point [YP]; moment the tissue changes its properties from elastic to plastic), strength (break point [BP]; moment of tissue disruption), and displacement at YP (marks the duration of the viscoelastic phase of the stretching) and BP (a measure of the strength of the material). Data were normalized to the dry weight of the cervix. RESULTS Hypoxia is associated with increased serum levels of VEGF compared to NP or NMX groups (P = .001). Cervical stiffness was lower in NMX, compared with NP animals (P = .004), and was not significantly influenced by hypoxia (P > .05). Overall there was a significant inverse correlation between slope and maternal serum levels of VEGF (r = -0.85, P < .001). The force required to reach YP was significantly higher for the NP, compared with NMX and HPX groups (P = .004). Hypoxia did not alter the force required to reach the YP (NMX vs HPX, P > .05). Conversely, hypoxia significantly decreased the displacement at YP, indicating a shortening of the elastic phase (NMX vs HPX, P = .021). There was a significant inverse correlation between maternal serum levels of VEGF and the displacement at YP (r = -0.68, P = .002). In vivo, hypoxia decreased the force required to reached the BP (NMX vs HPX, P = .025), but there was no correlation between the levels of maternal serum VEGF and this indicator (r = -0.35, P = .170). CONCLUSION Chronic hypoxia induces measurable changes in maternal serum levels of VEGF and tensile properties of the rat cervix, specifically a shortening of the elastic phase. Hypoxia decreases the cervical strength to stretch and predisposes to rupture, but this effect seems to be unrelated to maternal serum levels of VEGF.
Collapse
|
29
|
Gilbert JS, Babcock SA, Granger JP. Hypertension produced by reduced uterine perfusion in pregnant rats is associated with increased soluble fms-like tyrosine kinase-1 expression. Hypertension 2007; 50:1142-7. [PMID: 17923588 DOI: 10.1161/hypertensionaha.107.096594] [Citation(s) in RCA: 245] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The balance between proangiogenic and antiangiogenic factors, such as vascular endothelial growth factor, placental growth factor, and soluble fms-like tyrosine kinase-1 (sFlt-1), is altered in preeclampsia, and this dysregulation of angiogenic factors may be important in the pathogenesis of preeclampsia. Although sFlt-1 is elevated in preeclampsia, the mechanisms responsible for increasing this antiangiogenic factor remain unclear. We hypothesized that the hypertension produced by reduced uterine perfusion pressure (RUPP) is associated with increased sFlt-1 expression and decreased plasma vascular endothelial growth factor and placental growth factor concentrations in the pregnant rat. Arterial pressure was increased (130+/-3 versus 100+/-2 mm Hg; P<0.01) in the RUPP rats compared with the normal pregnant control rats. Plasma sFlt-1 concentration (660+/-270 versus 82+/-26 pg/mL; P<0.05) was increased, whereas plasma free placental growth factor (0.28+/-0.05 versus 1.7+/-0.5 pg/mL; P<0.01) and vascular endothelial growth factor (594+/-34 versus 830+/-33 pg/mL; P<0.01) concentrations were decreased in the RUPP rats compared with normal pregnant rats. Plasma sFlt-1:placental growth factor (37.2+/-7.8 versus 8.9+/-1.6; P<0.02) and sFlt-1:vascular endothelial growth factor (0.86+/-0.22 versus 0.28+/-0.06; P<0.05) ratios were increased in the RUPP rats compared with normal pregnant rats. Immunoreactive placental sFlt-1 was increased (1.1+/-0.1 versus 0.3+/-0.1; P<0.01) in RUPP rats contrasted with the normal pregnant rats. These findings support our hypothesis that RUPP increases the expression of sFlt-1 and alters the balance of angiogenic factors in the maternal circulation. These data also indicate that the RUPP model of pregnancy-induced hypertension may provide an invaluable model for mechanistic studies into the role of sFlt-1 in the pathogenesis preeclampsia.
Collapse
Affiliation(s)
- Jeffrey S Gilbert
- Department of Physiology and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA
| | | | | |
Collapse
|