Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1866(2): 165558, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31654701

ABSTRACT

Maternal pregestational obesity is a well-known risk factor for offspring obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes. The mechanisms by which maternal obesity can induce alterations in fetal and later neonatal metabolism are not fully elucidated due to its complexity and multifactorial causes. Two adipokines, leptin and adiponectin, are involved in fetal and postnatal growth trajectories, and both are altered in women with pregestational obesity. The placenta synthesizes leptin, which goes mainly to the maternal circulation and in lesser amount to the developing fetus. Maternal pregestational obesity and hyperleptinemia are associated with placental dysfunction and changes in nutrient transporters which directly affect fetal growth and development. By the other side, the embryo can produce its own leptin from early in development, which is associated to fetal weight and adiposity. Adiponectin, an insulin-sensitizing adipokine, is downregulated in maternal obesity. High molecular weight (HMW) adiponectin is the most abundant form and with most biological actions. In maternal obesity lower total and HMW adiponectin levels have been described in the mother, paralleled with high levels in the umbilical cord. Several studies have found that cord blood adiponectin levels are related with postnatal growth trajectories, and it has been suggested that low adiponectin levels in women with pregestational obesity enhance placental insulin sensitivity and activation of placental amino acid transport systems, supporting fetal overgrowth. The possible mechanisms by which maternal pregestational obesity, focusing in the actions of leptin and adiponectin, affects the fetal development and postnatal growth trajectories in their offspring are discussed.


Subject(s)
Adipokines/metabolism , Diabetes, Gestational/metabolism , Obesity, Maternal/metabolism , Adiponectin/metabolism , Female , Fetal Development/physiology , Humans , Insulin Resistance , Leptin/metabolism , Molecular Weight , Placenta/metabolism , Pregnancy , Pregnancy Complications/metabolism , Receptors, Adiponectin/metabolism , Receptors, Leptin/metabolism
2.
J Dev Orig Health Dis ; 7(3): 237-243, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26490762

ABSTRACT

In humans, obesity before and during pregnancy is associated with both fetal macrosomia and growth restriction, and long-term cardiovascular risk in the offspring. We aimed to determine whether overweighted pregnant guinea pig sows results in an increased fetal weight at term and the effects on the vascular reactivity in fetal systemic and umbilical arteries. Pregnant guinea pigs were classified as control (n=4) or high weight (HWS, n=5) according to their pre-mating weight, and their fetuses extracted at 0.9 gestation (~60 days). Segments of fetal femoral and umbilical arteries were mounted in a wire myograph, where the contractile response to KCl (5-125 mM), and the relaxation to nitric oxide synthase-dependent agents (insulin, 10-10-10-7 and acetylcholine, 10-10-10-5) and nitric oxide [sodium nitroprusside (SNP), 10-10-10-5] were determined. Fetuses from HWS (HWSF) were grouped according to their body weight as low (85 g) fetal weight, based on the confidence interval (76.5-84.9 g) of the control group. No HWSF were observed in the normal range. Umbilical arteries from HWSF showed a lower response to KCl and insulin compared with controls, but a comparable response with SNP. Conversely, femoral arteries from HWSF showed an increased response to KCl and acetylcholine, along with a decreased sensitivity to SNP. These data show that overweight sows have altered fetal growth along gestation. Further, large and small fetuses from obese guinea pig sows showed altered vascular reactivity at umbilical and systemic vessels, which potentially associates with long-term cardiovascular risk.

3.
Placenta ; 36(5): 552-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25747728

ABSTRACT

INTRODUCTION: Fetal macrosomia and intrauterine growth restriction (IUGR) associate with increased morbidity in the neonate. Placental vascular relaxation is impaired in fetal macrosomia, as well as in IUGR, and this could result from increased oxidative stress present in both conditions. We determined the role of pro- and anti-oxidants on NOS dependent relaxation in placental chorionic arteries from pregnancies with LGA babies from overweight and/or obese mothers (LOOM) and IUGR fetuses from normal BMI women. METHODS: Chorionic arteries were mounted in a wire-myograph, where responses to the NOS-dependent agent CGRP in presence or absence of the antioxidant N-acetyl cysteine (NAC), the pro-oxidant SIN-1, the SOD inhibitor DDC, and the GPx inhibitor MS were determined. Additionally the presence of pro- and antioxidant enzymes (NOX-4, SOD-1, SOD-2 and GPx-1) and eNOS in chorionic and umbilical vessels were addressed by immunohistochemistry. RESULTS: Maximal CGRP-induced relaxation was comparable to controls but presented a reduced potency in chorionic arteries from LOOM placentae, whilst in IUGR vessels both maximal response and potency were reduced. NAC increased maximal relaxation in controls, IUGR and LOOM arteries, whilst SIN-1 completely abolished the CGRP-induced relaxation only in IUGR and LOOM samples, the later effect was paralleled by SOD or GPx inhibition. These responses associated with the presence of NOX-4, SOD-1 and GPx-1 in the endothelium and vascular wall of chorionic and umbilical arteries in the different groups studied. DISCUSSION: These data suggest that NOS dependent relaxation in placental vessels from IUGR and LOOM pregnancies present a higher sensitivity to oxidative stress.


Subject(s)
Arteries/physiopathology , Endothelium, Vascular/physiopathology , Fetal Growth Retardation/physiopathology , Fetal Macrosomia/physiopathology , Obesity/physiopathology , Adult , Antioxidants/metabolism , Arteries/metabolism , Case-Control Studies , Female , Glutathione Peroxidase/metabolism , Humans , In Vitro Techniques , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Placenta/physiopathology , Pregnancy , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Glutathione Peroxidase GPX1
4.
J Neuroendocrinol ; 26(9): 587-602, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24690036

ABSTRACT

This review discusses the effects of stress and nutrition throughout development and summarises studies investigating how exposure to stress or alterations in nutrition during the pre-conception, prenatal and early postnatal periods can affect the long-term health of an individual. In general, the data presented here suggest that that anything signalling potential adverse conditions later in life, such as high levels of stress or low levels of food availability, will lead to alterations in the offspring, possibly of an epigenetic nature, preparing the offspring for these conditions later in life. However, when similar environmental conditions are not met in adulthood, these alterations may have maladaptive consequences, resulting in obesity and heightened stress sensitivity. The data also suggest that the mechanism underlying these adult phenotypes might be dependent on the type and the timing of exposure.


Subject(s)
Child Nutritional Physiological Phenomena/physiology , Maternal Nutritional Physiological Phenomena/physiology , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/physiopathology , Animals , Brain/growth & development , Child , Child Nutritional Physiological Phenomena/genetics , Diet/adverse effects , Epigenesis, Genetic , Female , Glucocorticoids/physiology , Humans , Infant, Newborn , Male , Maternal Nutritional Physiological Phenomena/genetics , Obesity/etiology , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Stress, Psychological/genetics
5.
BJOG ; 120 Suppl 2: 3-8, v, 2013 09.
Article in English | MEDLINE | ID: mdl-23678966

ABSTRACT

BACKGROUND: Healthy growth in utero and after birth is fundamental for lifelong health and wellbeing. The World Health Organization (WHO) recently published standards for healthy growth from birth to 6 years of age; analogous standards for healthy fetal growth are not currently available. Current fetal growth charts in use are not true standards, since they are based on cross-sectional measurements of attained size under conditions that do not accurately reflect normal growth. In most cases, the pregnant populations and environments studied are far from ideal; thus the data are unlikely to reflect optimal fetal growth. A true standard should reflect how fetuses and newborns 'should' grow under ideal environmental conditions. OBJECTIVE: The development of prescriptive intrauterine and newborn growth standards derived from the INTERGROWTH-21(st) Project provides the data that will allow us for the first time to establish what is 'normal' fetal growth. METHODS: The INTERGROWTH-21(st) study centres provide the data set obtained under pre-established standardised criteria, and details of the methods used are also published. DESIGN: Multicentre study with sites in all major geographical regions of the world using a standard evaluation protocol. RESULTS: These standards will assess risk of abnormal size at birth and serve to evaluate potentially effective interventions to promote optimal growth beyond securing survival. DISCUSSION: The new normative standards have the potential to impact perinatal and neonatal survival and beyond, particularly in developing countries where fetal growth restriction is most prevalent. They will help us identify intrauterine growth restriction at earlier stages of development, when preventive or corrective strategies might be more effective than at present. CONCLUSION: These growth standards will take us one step closer to effective action in preventing and potentially reversing abnormal intrauterine growth. Achieving 'optimal' fetal growth requires that we act not only during pregnancy but that we optimize the maternal uterine environment from the time before conception, through embryonic development until fetal growth is complete. The remaining challenge is how 'early' will we be able to act, now that we can better monitor fetal growth.


Subject(s)
Child Development , Fetal Development , Growth Charts , Infant, Newborn/growth & development , Multicenter Studies as Topic , Child , Child, Preschool , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/etiology , Fetal Growth Retardation/prevention & control , Humans , Infant , Infant, Low Birth Weight/growth & development , Infant, Premature/growth & development , Multicenter Studies as Topic/methods , Multicenter Studies as Topic/standards , Pregnancy
6.
Placenta ; 34(1): 20-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23122700

ABSTRACT

Placental vascular tone is critically influenced by nitric oxide (NO) derived from endothelial NO synthase (eNOS) activity. Placental vessels from pregnancies complicated with intrauterine growth restriction present altered NOS-dependent vasodilation. Arginase-2 competes with eNOS for l-arginine and counteracts the NOS-dependent relaxation in umbilical vessels from normal pregnancies. However there is no data regarding the contribution of arginase activity on the impaired endothelial function in IUGR placenta. We studied whether arginase-2 participates in IUGR-related placental vascular dysfunction counteracting eNOS-dependent relaxation, and the regulation of arginase-2 and eNOS expression in endothelial cells from IUGR umbilical arteries (HUAEC) and veins (HUVEC). In IUGR-derived umbilical arteries (UA) and veins (UV), and chorionic arteries (CA), NOS-dependent vasoactive response in the presence and absence of BEC (arginase inhibitor) was studied. Protein levels of eNOS (total and Ser(1177)-P-eNOS), arginase-2 and arginase activity were determined in IUGR HUAEC and HUVEC. In IUGR vessels eNOS-dependent relaxation was reduced, being improved by BEC. This effect was higher in arteries than veins, and in chorionic compared with umbilical vessels. In cultured IUGR endothelial cells, arginase-2 protein expression and activity were increased in HUVEC, without changes in HUAEC. In IUGR-derived endothelium there was a generalized reduction in the in vitro eNOS activation (Ser(1177)-P-eNOS/eNOS), and therefore a decreased eNOS/arginase activity ratio. Here we provide ex vivo and in vitro evidence for a vascular role of arginase throughout placental vasculature, negatively controlling NOS activity. This effect seems to be crucial in the pathophysiology of endothelial dysfunction present in IUGR feto-placental vessels.


Subject(s)
Arginase/metabolism , Blood Vessels/physiopathology , Endothelial Cells/metabolism , Fetal Growth Retardation/physiopathology , Nitric Oxide Synthase Type III/metabolism , Placenta/blood supply , Umbilical Arteries/physiopathology , Adult , Arginase/physiology , Blood Vessels/metabolism , Blood Vessels/pathology , Cells, Cultured , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Infant, Newborn , Male , Nitric Oxide Synthase Type III/physiology , Placenta/metabolism , Placenta/pathology , Placental Circulation/physiology , Pregnancy , Umbilical Arteries/metabolism , Umbilical Arteries/pathology
7.
Placenta ; 33(5): 360-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22391327

ABSTRACT

The main vasodilator in the placenta is nitric oxide (NO), which is synthesized by endothelial NO synthase (eNOS). Arginase-2 competes with eNOS for l-arginine, and its activity has been related with vascular dysfunction. Recently, we showed that hypoxia induces arginase-2, and decreases eNOS activity in human umbilical vein endothelial cells (HUVEC). However there is evidence that vascular responses to hypoxia are not similar throughout the placental vascular tree. We studied whether arginase-2 plays a role controlling vascular tone in human umbilical vessels, and the changes in the expression of arginase-2 and eNOS proteins by hypoxia in endothelial cells from umbilical arteries (HUAEC) and veins (HUVEC). In isolated umbilical vessels the presence of eNOS and arginase-2 was determined in the endothelium, and the NO-dependent vasoactive responses in the presence and absence of S-(2-boronoethyl)-L-cysteine (BEC, arginase inhibitor) were studied. Additionally, HUAEC and HUVEC were exposed (0-24 h) to hypoxia (2% O2) or normoxia (5% O2), and protein levels of eNOS (total and phosphorylated at serine-1177) and arginase-2 were determined. In umbilical arteries and veins arginase-2 and eNOS were detected mainly at the endothelium. BEC induced a higher concentration-dependent relaxation in umbilical arteries than veins, and these responses were NOS-dependent. In HUAEC exposed to hypoxia there were no changes in eNOS and arginase-2 levels, however there was a significant increase of p-eNOS. In contrast, HUVEC showed an increase in arginase-2 and a reduction of p-eNOS in response to hypoxia. These results show that arginases have a vascular role in placental vessels counteracting the NOS-dependent relaxation, which is differentially regulated in placental artery and vein endothelial cells.


Subject(s)
Arginase/metabolism , Nitric Oxide Synthase Type III/metabolism , Pregnancy/metabolism , Umbilical Arteries/enzymology , Umbilical Veins/enzymology , Vasodilation , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Female , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia/enzymology , Myography
8.
Placenta ; 32(12): 932-40, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21962305

ABSTRACT

Hypoxia relates with altered placental vasodilation, and in isolated endothelial cells, it reduces activity of the endothelial nitric oxide synthase (eNOS) and l-arginine transport. It has been reported that arginase-2 expression, an alternative pathway for l-arginine metabolism, is increased in adult endothelial cells exposed to hypoxia as well as in pre-eclamptic placentae. We studied in human umbilical vein endothelial cells (HUVEC) whether hypoxia-reduced NO synthesis results from increased arginase-mediated l-arginine metabolism and changes in subcellular localization of eNOS and arginase-2. In HUVEC exposed (24 h) to 5% (normoxia) or 2% (hypoxia) oxygen, l-arginine transport kinetics, arginase activity (urea assay), and NO synthase (NOS) activity (l-citrulline assay) were determined. Arginase-1, arginase-2 and eNOS expression were determined by RT-PCR and Western blot. Subcellular localization of arginase-2 and eNOS were studied using confocal microscopy and indirect immunofluorescence. Experiments were done in absence or presence of S-(2-boronoethyl)-l-cysteine-HCl (BEC, arginase inhibitor) or N(G)-nitro-l-arginine methyl ester (l-NAME). Hypoxia-induced reduction in eNOS activity was associated with a reduction in eNOS phosphorylation at Serine-1177 and increased phosphorylation at Threonine-495. This was paralleled with an induction in arginase-2 expression and activity, and decreased l-arginine transport. In hypoxia the arginase inhibition, restored NO synthesis and l-arginine transport, without changes in the eNOS post-translational modification status. Hypoxia increased arginase-2/eNOS colocalization, and eNOS redistribution to the cell periphery. Altogether these data reinforce the thought that eNOS cell location, post-translational modification and substrate availability are important mechanisms regulating eNOS activity. If these mechanisms occur in pregnancy diseases where feto-placental oxygen levels are reduced remains to be clarified.


Subject(s)
Arginase/biosynthesis , Human Umbilical Vein Endothelial Cells/enzymology , Hypoxia/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Adult , Arginase/antagonists & inhibitors , Arginine/metabolism , Boronic Acids/pharmacology , Female , Fluorescent Antibody Technique, Indirect , Humans , Microscopy, Confocal , NG-Nitroarginine Methyl Ester/pharmacology , Phosphorylation , Pregnancy , Protein Processing, Post-Translational , Subcellular Fractions/enzymology
9.
Placenta ; 32(11): 797-805, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21798594

ABSTRACT

Nitric oxide (NO) is one of the most pleiotropic signaling molecules at systemic and cellular levels, participating in vascular tone regulation, cellular respiration, proliferation, apoptosis and gene expression. Indeed NO actively participates in trophoblast invasion, placental development and represents the main vasodilator in this tissue. Despite the large number of studies addressing the role of NO in the placenta, its participation in placental vascular development and the effect of altered levels of NO on placental function remains to be clarified. This review draws a time-line of the participation of NO throughout placental vascular development, from the differentiation of vascular precursors to the consolidation of vascular function are considered. The influence of NO on cell types involved in the origin of the placental vasculature and the expression and function of the nitric oxide synthases (NOS) throughout pregnancy are described. The developmental processes involved in the placental vascular bed are considered, such as the participation of NO in placental vasculogenesis and angiogenesis through VEGF and Angiopoietin signaling molecules. The role of NO in vascular function once the placental vascular tree has developed, in normal pregnancy as well as in pregnancy-related diseases, is then discussed.


Subject(s)
Neovascularization, Physiologic/physiology , Nitric Oxide/physiology , Placenta/blood supply , Animals , Female , Humans , Models, Biological , Neovascularization, Physiologic/genetics , Nitric Oxide/metabolism , Placenta/metabolism , Placenta/physiology , Placenta Diseases/etiology , Placenta Diseases/genetics , Placenta Diseases/metabolism , Placentation/genetics , Placentation/physiology , Pregnancy , Pregnancy Complications/etiology , Pregnancy Complications/metabolism
10.
Placenta ; 32 Suppl 2: S159-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21215450

ABSTRACT

Human endothelial dysfunction is a common feature in many diseases of pregnancy, such as gestational diabetes (GD). Metabolic changes include abnormal synthesis of nitric oxide (NO) and abnormal membrane transport of l-arginine and adenosine in primary cultures of human umbilical vein (HUVEC, macrovascular) and placental microvillus (hPMEC, microvascular) endothelial cells. These alterations are associated with modifications in the expression and activity of endothelial (eNOS) and inducible (iNOS) NO synthases, respectively, an effect that is maintained at least up to passage 5 in culture. HUVEC and hPMEC exhibit expression and activity of the human cationic amino acid transporter 1 (hCAT-1), equilibrative nucleoside transporters 1 (hENT1) and hENT2, as well as the corresponding SLC7A1, SLC29A1 and SLC29A2 gene promoter activities. Altered gene expression results from increased NO level, protein kinase C, mitogen-activated protein kinases, and hCHOP-C/EBPα transcription factor activation. Reduced ENT-mediated adenosine transport in GD is associated with stimulation of the l-arginine/NO pathway, and mainly due to reduced expression and activity of hENT1. In addition, hENT2 activity seems able to restore the reduced adenosine transport in GD. Additionally, insulin exerts a differential modulation of endothelial cells from macrocirculation compared with microcirculation, possibly due to expression of different insulin receptor isoforms. It is suggested that a common functional characteristic leading to changes in the bioavailability of adenosine and metabolism of l-arginine is evidenced by human fetal micro and macrovascular endothelium in GD.


Subject(s)
Diabetes, Gestational/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Microvessels/physiopathology , Placenta/blood supply , Placenta/physiopathology , Diabetes, Gestational/metabolism , Endothelium, Vascular/cytology , Female , Humans , Microvessels/cytology , Microvessels/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy
11.
Placenta ; 30(7): 625-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19501907

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with chronic fetal hypoxia, altered placental vasodilatation and reduced endothelial nitric oxide synthase (eNOS) activity. In human umbilical vein endothelial cells (HUVEC) from pregnancies complicated with IUGR (IUGR cells) and in HUVEC from normal pregnancies (normal cells) cultured under hypoxia l-arginine transport is reduced; however, the mechanisms leading to this dysfunction are unknown. We studied hypoxia effect on l-arginine transport and human cationic amino acid transporters 1 (hCAT-1) expression, and the potential NO and protein kinase C alpha (PKCalpha) involvement. Normal or IUGR HUVEC monolayers were exposed (0-24h) to 5% O(2) (normoxia), and 1 or 2% O(2) (hypoxia). l-Arginine transport and hCAT-1 expression, phosphorylated and total PKCalpha or eNOS protein and mRNA expression were quantified. eNOS involvement was tested using a siRNA against eNOS (eNOS-siRNA) adenovirus. IUGR cells in normoxia or hypoxia, and normal cells in hypoxia exhibited reduced l-arginine transport, hCAT-1 expression, NO synthesis and eNOS phosphorylation at Serine(1177), effects reversed by calphostin C (PKC inhibitor) and S-nitroso-N-acetyl-l,d-penicillamine (SNAP, NO donor). However, N(G)-nitro-l-arginine methyl ester (l-NAME, NOS inhibitor) reduced hCAT-1 expression only in normal cells in normoxia. Increased Thr(638)-phosphorylated PKCalpha was exhibited by IUGR cells in normoxia or hypoxia and normal cells in hypoxia. The effects of hypoxia in normal cells were mimicked in eNOS-siRNA transduced cells; however, IUGR phenotype was unaltered by eNOS knockdown. Thus, IUGR- and hypoxia-reduced l-arginine transport could result from increased PKCalpha, but reduced eNOS activity leading to a lower hCAT-1 expression in HUVEC. In addition, IUGR endothelial cells are either not responsive or maximally affected by hypoxia. These mechanisms could be responsible for placental dysfunction in diseases where fetal endothelium is chronically exposed to hypoxia, such as IUGR.


Subject(s)
Arginine/metabolism , Endothelial Cells/enzymology , Fetal Growth Retardation/metabolism , Hypoxia/metabolism , Nitric Oxide/biosynthesis , Umbilical Veins/cytology , Adult , Cationic Amino Acid Transporter 1/genetics , Cationic Amino Acid Transporter 1/metabolism , Cells, Cultured , Endothelial Cells/cytology , Female , Humans , Models, Biological , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/physiology , Pregnancy , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , RNA, Messenger/metabolism , Vasodilation/physiology , Young Adult
12.
Placenta ; 29(9): 816-25, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18703227

ABSTRACT

Pre-eclampsia is associated with elevated maternal blood pressure and proteinuria, altered fetal growth, and increased plasma adenosine concentration in the mother and the fetus. Human equilibrative nucleoside transporters 1 (hENT1) and hENT2 are crucial to maintain physiological plasma levels of adenosine, thus modulating its several biological effects through adenosine receptor activation. However, it is unknown whether hENTs and adenosine receptors are expressed in human placental microvascular endothelium (hPMEC). To assay whether the increased fetal plasma adenosine concentration in pre-eclampsia results from altered hENT-mediated transport, and the potential involvement of adenosine receptors in this phenomenon, we investigated hENTs and A2A and A2B adenosine receptors expression and function in hPMEC. Cells were isolated and cultured from normal pregnancies (n=17) or pre-eclampsia with adequate-for-gestational age fetuses (n=7). hENT1, hENT2, A2A and A2B adenosine receptors were expressed and functional in hPMEC. Extracellular adenosine concentration was higher (4-fold) in pre-eclampsia versus normal pregnancies. hPMEC from pre-eclampsia exhibit increased total transport (hENT1+hENT2), and maximal velocity (Vmax) for hENT2- (2-fold), but reduced Vmax for hENT1-mediated adenosine transport (75%), with no changes in apparent Km. hENT2 expression was increased (4.5-fold), but hENT1 protein abundance was reduced (80%) in pre-eclampsia. Equally, A2A expression was reduced (50-80%) in pre-eclampsia. CGS-21680 (A2A agonist) did not alter hENTs expression or activity, but ZM-241385 (A2A antagonist) blocked pre-eclampsia effects and increased hENT1-mediated transport in normal pregnancies. Thus, A2B adenosine receptors may differentially modulate hENTs in hPMEC, which could be a mechanism attempting to re-establish physiological extracellular adenosine levels in pre-eclampsia.


Subject(s)
Endothelial Cells/physiology , Equilibrative Nucleoside Transporter 1/biosynthesis , Equilibrative-Nucleoside Transporter 2/biosynthesis , Placenta/cytology , Pre-Eclampsia/physiopathology , Receptor, Adenosine A2B/physiology , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Adult , Equilibrative Nucleoside Transporter 1/physiology , Equilibrative-Nucleoside Transporter 2/physiology , Female , Gene Expression Regulation , Humans , Phenethylamines/pharmacology , Pregnancy , Receptor, Adenosine A2A/biosynthesis , Triazines/pharmacology , Triazoles/pharmacology
13.
Placenta ; 26(8-9): 641-53, 2005.
Article in English | MEDLINE | ID: mdl-16085043

ABSTRACT

Human equilibrative, Na(+)-independent nucleoside transport is mediated by membrane proteins sensitive (system es, hENT1) or insensitive (system ei, hENT2) to nitrobenzylthioinosine (NBMPR). Gestational diabetes and elevated extracellular concentrations of D-glucose reduce adenosine transport in human umbilical vein endothelium (HUVEC). We studied hENT2 and hENT1 expression in HUVEC, and the effect of D-glucose on their activity and expression in HUVEC preincubated with 25 mM D-glucose (24 h). hENT2 and hENT1 mRNA were quantified by real-time reverse transcription polymerase chain reaction, and their proteins were detected by Western blotting. hENT2 and hENT1 proteins are co-expressed in HUVEC and are located at the plasma membrane, however, hENT2 was mainly cytoplasmatic and perinuclear in location. D-Glucose reduced hENT1 and hENT2 mRNA expression, but only hENT1 protein abundance at the plasma membrane. Adenosine transport was inhibited by D-glucose and NMBPR (1 microM) in intact cells and membrane vesicles. Hypoxanthine inhibited adenosine transport in the absence or in the presence of 1 microM NBMPR. D-Glucose reduced NBMPR maximal binding in intact cells, membrane vesicles, and plasma membrane fractions. In conclusion, the present study demonstrates that hENT2 and hENT1 are co-expressed in HUVEC, and even when adenosine transport is also mediated by hENT2, the hENT2-mediated transport activity is not involved in the d-glucose-induced down-regulation of total adenosine transport.


Subject(s)
Adenosine/metabolism , Endothelium, Vascular/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Hypoglycemia/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative-Nucleoside Transporter 2/genetics , Glucose/pharmacology , Humans , Hypoxanthine/pharmacology , Nucleoside Transport Proteins/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...