Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(1): e0245155, 2021.
Article in English | MEDLINE | ID: mdl-33434211

ABSTRACT

INTRODUCTION: Impaired utero-placental perfusion is a well-known feature of early preeclampsia and is associated with placental hypoxia and oxidative stress. Although aberrations at the level of the mitochondrion have been implicated in PE pathophysiology, whether or not hypoxia-induced mitochondrial abnormalities contribute to placental oxidative stress is unknown. METHODS: We explored whether abnormalities in mitochondrial metabolism contribute to hypoxia-induced placental oxidative stress by using both healthy term placentae as well as a trophoblast cell line (BeWo cells) exposed to hypoxia. Furthermore, we explored the therapeutic potential of the antioxidants MitoQ and quercetin in preventing hypoxia-induced placental oxidative stress. RESULTS: Both in placental explants as well as BeWo cells, hypoxia resulted in reductions in mitochondrial content, decreased abundance of key molecules involved in the electron transport chain and increased expression and activity of glycolytic enzymes. Furthermore, expression levels of key regulators of mitochondrial biogenesis were decreased while the abundance of constituents of the mitophagy, autophagy and mitochondrial fission machinery was increased in response to hypoxia. In addition, placental hypoxia was associated with increased oxidative stress, inflammation, and apoptosis. Moreover, experiments with MitoQ revealed that hypoxia-induced reactive oxygen species originated from the mitochondria in the trophoblasts. DISCUSSION: This study is the first to demonstrate that placental hypoxia is associated with mitochondrial-generated reactive oxygen species and significant alterations in the molecular pathways controlling mitochondrial content and function. Furthermore, our data indicate that targeting mitochondrial oxidative stress may have therapeutic benefit in the management of pathologies related to placental hypoxia.


Subject(s)
Mitochondria/metabolism , Organelle Biogenesis , Oxidative Stress , Pre-Eclampsia/metabolism , Reactive Oxygen Species/metabolism , Trophoblasts/metabolism , Cell Hypoxia , Cell Line , Female , Humans , Mitochondria/pathology , Pre-Eclampsia/pathology , Pregnancy , Trophoblasts/pathology
2.
PLoS Biol ; 18(11): e3000885, 2020 11.
Article in English | MEDLINE | ID: mdl-33170835

ABSTRACT

Hypertension is the most important cause of death and disability in the elderly. In 9 out of 10 cases, the molecular cause, however, is unknown. One mechanistic hypothesis involves impaired endothelium-dependent vasodilation through reactive oxygen species (ROS) formation. Indeed, ROS forming NADPH oxidase (Nox) genes associate with hypertension, yet target validation has been negative. We re-investigate this association by molecular network analysis and identify NOX5, not present in rodents, as a sole neighbor to human vasodilatory endothelial nitric oxide (NO) signaling. In hypertensive patients, endothelial microparticles indeed contained higher levels of NOX5-but not NOX1, NOX2, or NOX4-with a bimodal distribution correlating with disease severity. Mechanistically, mice expressing human Nox5 in endothelial cells developed-upon aging-severe systolic hypertension and impaired endothelium-dependent vasodilation due to uncoupled NO synthase (NOS). We conclude that NOX5-induced uncoupling of endothelial NOS is a causal mechanism and theragnostic target of an age-related hypertension endotype. Nox5 knock-in (KI) mice represent the first mechanism-based animal model of hypertension.


Subject(s)
Hypertension/physiopathology , NADPH Oxidase 5/genetics , Nitric Oxide/metabolism , Adult , Age Factors , Aged , Animals , Endothelial Cells , Endothelium, Vascular , Female , Gene Knock-In Techniques/methods , Humans , Hypertension/genetics , Hypertension/metabolism , Male , Membrane Proteins/genetics , Mice , Middle Aged , NADPH Oxidase 5/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species
3.
Hypertens Res ; 43(12): 1361-1374, 2020 12.
Article in English | MEDLINE | ID: mdl-32733105

ABSTRACT

Preeclampsia (PE) is a pregnancy-related disorder characterized by hypertension and proteinuria that affects 3-10% of all pregnancies. Although its pathophysiology remains obscure, placental hypoxia-induced oxidative stress and alterations in vascular function, morphology, and endothelial barrier integrity are considered to play a key role in the development of preeclampsia. In this study, placental villous explants of noncomplicated placentae and BeWo cells were subjected to hypoxia. The effect of placental hypoxic-conditioned medium (HCM) on intraluminal-induced contraction and endothelial barrier integrity in chorionic arteries was investigated using pressure myography. The impact of BeWo cell HCM on endothelial cell viability, reactive oxygen species formation and inflammation was also determined. Alterations in arterial morphology and contractile responsiveness to the thromboxane A2 analog (U46619) after exposure to placental HCM were examined immunohistochemically and by wire myography, respectively. Intraluminal administration of placental HCM induced vasoconstriction and increased the endothelial permeability for KCl, which was concentration-dependently prevented by quercetin. Placental and BeWo cell HCMs decreased endothelial cell viability, increased the production of reactive oxygen species and enhanced the secretion of IL-6 and IL-8. The cross-sectional area of the arterial media was increased upon exposure to placental HCM, which was associated with increased vascular proliferation and contractile responsiveness to U46619, and all of these effects were prevented by the antioxidants quercetin and RRR-α-tocopherol. This study is the first to comprehensively demonstrate the link between factors secreted by placental cells in response to hypoxia and vascular abnormalities and paves the way for new diagnostic approaches and therapies to better protect the maternal vasculature during and after a preeclampsia-complicated pregnancy.


Subject(s)
Endothelium, Vascular/physiopathology , Hypoxia/physiopathology , Placenta/physiopathology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Cell Line , Culture Media, Conditioned , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Female , Humans , In Vitro Techniques , Permeability , Placenta/blood supply , Pre-Eclampsia/etiology , Pregnancy , Reactive Oxygen Species/metabolism , Vasoconstriction
4.
PLoS One ; 13(8): e0202648, 2018.
Article in English | MEDLINE | ID: mdl-30142162

ABSTRACT

INTRODUCTION: Preeclampsia is a major health problem in human pregnancy, severely complicating 5-8% of all pregnancies. The emerging molecular mechanism is that conditions like hypoxic stress trigger the release of placental messengers into the maternal circulation, which causes preeclampsia. Our objective was to develop an in vitro model, which can be used to further elucidate the molecular mechanisms of preeclampsia and which might be used to find a remedy. METHODS: Human non-complicated term placentas were collected. Placental explants were subjected to severe hypoxia and the conditioned media were added to chorionic arteries that were mounted into a myograph. Contractile responses of the conditioned media were determined, as well as effects on thromboxane-A2 (U46619) induced contractility. To identify the vasoactive compounds present in the conditioned media, specific receptor antagonists were evaluated. RESULTS: Factors released by placental explants generated under severe hypoxia induced an increased vasoconstriction and vascular contractility to thromboxane-A2. It was found that agonists for the angiotensin-I and endothelin-1 receptor released by placental tissue under severe hypoxia provoke vasoconstriction. The dietary antioxidant quercetin could partially prevent the acute and sustained vascular effects in a concentration-dependent manner. DISCUSSION: Both the acute vasoconstriction, as well as the increased contractility to U46619 are in line with the clinical vascular complications observed in preeclampsia. Data obtained with quercetin supports that our model opens avenues for e.g. nutritional interventions aimed at treating or preventing preeclampsia.


Subject(s)
Constriction, Pathologic/genetics , Placenta/metabolism , Pre-Eclampsia/genetics , Vasoconstriction/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Chorion/blood supply , Chorion/metabolism , Chorion/pathology , Constriction, Pathologic/physiopathology , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Hypoxia/genetics , Hypoxia/pathology , Muscle Contraction/drug effects , Myography , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
5.
Biochim Biophys Acta Gen Subj ; 1861(5 Pt A): 1177-1189, 2017 May.
Article in English | MEDLINE | ID: mdl-28286015

ABSTRACT

BACKGROUND: Rutin intake is associated with a reduced risk of cardiovascular disease (CVD). The exact mechanism by which rutin can protect against CVD development is still enigmatic. Since, rutin is a compound with a relatively short half-life, the direct antioxidant effect of rutin cannot explain the long-lasting effect on human health. We hypothesized that rutin next to its direct antioxidant effect that improves endothelial function, may also induce an adaptive response in endogenous antioxidant systems. METHODS AND RESULTS: In Human Umbilical Vein Endothelial Cells (HUVECs), the direct antioxidant effect was confirmed. During scavenging of Reactive Oxygen Species (ROS), rutin is oxidized into a quinone derivative. HUVECs pretreated with rutin quinone became better protected against a second challenge with oxidative stress 3h later. LC-MS/MS analysis indicated that rutin quinone targets cysteine 151 of Keap1. Moreover, we found that the quinone is an inhibitor of the selenoprotein thioredoxin reductase 1. These properties correlated with an activation of Nrf2 and upregulation of Glutamate Cysteine Ligase, the rate-limiting enzyme of glutathione synthesis, while NF-κB and HIF activation became blunted by rutin treatment. Furthermore, rutin was found to prevent hydrogen peroxide from impairing relaxation of human chorionic plate placental vessels, which may help to protect endothelial function. CONCLUSION AND SIGNIFICANCE: Rutin functions as an antioxidant and is oxidized into a quinone that upregulates the Nrf2-mediated endogenous antioxidant response. This mechanism suggests that rutin selectively exerts its protective effects in regions with increased oxidative stress, and explains how rutin reduces the risk of developing CVD. GENERAL SIGNIFICANCE: The newly found mechanism behind the long-term protection of rutin against cardiovascular disease, the selective upregulation of endogenous antioxidant systems, contributes to the further understanding why rutin can reduce the risk on developing cardiovascular disease.


Subject(s)
Adaptation, Physiological/drug effects , Arterioles/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rutin/pharmacology , Antioxidants/pharmacology , Arterioles/metabolism , Cells, Cultured , Female , Glutamate-Cysteine Ligase/metabolism , HEK293 Cells , Half-Life , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , NF-kappa B/metabolism , Oxidation-Reduction/drug effects , Placenta/drug effects , Placenta/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
6.
Clin Sci (Lond) ; 129(12): 1061-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26464516

ABSTRACT

Reduced nitric oxide (NO)/cGMP signalling is observed in age-related vascular disease. We hypothesize that this disturbed signalling involves effects of genomic instability, a primary causal factor in aging, on vascular smooth muscle cells (VSMCs) and that the underlying mechanism plays a role in human age-related vascular disease. To test our hypothesis, we combined experiments in mice with genomic instability resulting from the defective nucleotide excision repair gene ERCC1 (Ercc1(d/-) mice), human VSMC cultures and population genome-wide association studies (GWAS). Aortic rings of Ercc1(d/-) mice showed 43% reduced responses to the soluble guanylate cyclase (sGC) stimulator sodium nitroprusside (SNP). Inhibition of phosphodiesterase (PDE) 1 and 5 normalized SNP-relaxing effects in Ercc1(d/-) to wild-type (WT) levels. PDE1C levels were increased in lung and aorta. cGMP hydrolysis by PDE in lungs was higher in Ercc1(d/-) mice. No differences in activity or levels of cGMP-dependent protein kinase 1 or sGC were observed in Ercc1(d/-) mice compared with WT. Senescent human VSMC showed elevated PDE1A and PDE1C and PDE5 mRNA levels (11.6-, 9- and 2.3-fold respectively), which associated with markers of cellular senescence. Conversely, PDE1 inhibition lowered expression of these markers. Human genetic studies revealed significant associations of PDE1A single nucleotide polymorphisms with diastolic blood pressure (DBP; ß=0.28, P=2.47×10(-5)) and carotid intima-media thickness (cIMT; ß=-0.0061, P=2.89×10(-5)). In summary, these results show that genomic instability and cellular senescence in VSMCs increase PDE1 expression. This might play a role in aging-related loss of vasodilator function, VSMC senescence, increased blood pressure and vascular hypertrophy.


Subject(s)
Aging/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Vasodilation , Aging/genetics , Animals , Blood Pressure , Carotid Arteries/enzymology , Carotid Arteries/pathology , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Intima-Media Thickness , Cells, Cultured , Cellular Senescence , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Endonucleases/deficiency , Endonucleases/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hydrolysis , Hyperplasia , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , In Vitro Techniques , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phenotype , Phosphodiesterase 5 Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Second Messenger Systems , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
Circulation ; 122(3): 273-81, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20606119

ABSTRACT

BACKGROUND: Control of peripheral resistance arteries by autonomic nerves is essential for the regulation of blood flow. The signals responsible for the maintenance of vascular neuroeffector mechanisms in the adult, however, remain largely unknown. METHODS AND RESULTS: Here, we report that VEGF( partial differential/ partial differential) mice with low vascular endothelial growth factor (VEGF) levels suffer defects in the regulation of resistance arteries. These defects are due to dysfunction and structural remodeling of the neuroeffector junction, the equivalent of a synapse between autonomic nerve endings and vascular smooth muscle cells, and to an impaired contractile smooth muscle cell phenotype. Notably, short-term delivery of a VEGF inhibitor to healthy mice also resulted in functional and structural defects of neuroeffector junctions. CONCLUSIONS: These findings uncover a novel role for VEGF in the maintenance of arterial neuroeffector function and may help us better understand how VEGF inhibitors cause vascular regulation defects in cancer patients.


Subject(s)
Autonomic Nervous System Diseases/physiopathology , Cardiovascular Diseases/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Resistance/physiology , Vasoconstriction/physiology , Animals , Autonomic Nervous System Diseases/genetics , Cardiovascular Diseases/genetics , Carotid Artery, Common/innervation , Carotid Artery, Common/physiology , Gene Expression/physiology , Gene Transfer Techniques , Lac Operon , Mesenteric Arteries/innervation , Mesenteric Arteries/physiology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
J Pharmacol Exp Ther ; 333(1): 210-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20040579

ABSTRACT

We tested the hypothesis that changes in arterial blood flow modify the function of endothelial Ca2+-activated K+ channels [calcium-activated K+ channel (K(Ca)), small-conductance calcium-activated K+ channel (SK3), and intermediate calcium-activated K+ channel (IK1)] before arterial structural remodeling. In rats, mesenteric arteries were exposed to increased [+90%, high flow (HF)] or reduced blood flow [-90%, low flow (LF)] and analyzed 24 h later. There were no detectable changes in arterial structure or in expression level of endothelial nitric-oxide synthase, SK3, or IK1. Arterial relaxing responses to acetylcholine and 3-oxime-6,7-dichlore-1H-indole-2,3-dione (NS309; activator of SK3 and IK1) were measured in the absence and presence of endothelium, NO, and prostanoid blockers, and 6,12,19,20,25,26-hexahydro-5,27:13,18:21,24-trietheno-11,7-metheno-7H-dibenzo [b,n] [1,5,12,16]tetraazacyclotricosine-5,13-diium dibromide (UCL 1684; inhibitor of SK3) or 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34; inhibitor of IK1). In LF arteries, endothelium-dependent relaxation was markedly reduced, due to a reduction in the endothelium-derived hyperpolarizing factor (EDHF) response. In HF arteries, the balance between the NO/prostanoid versus EDHF response was unaltered. However, the contribution of IK1 to the EDHF response was enhanced, as indicated by a larger effect of TRAM-34 and a larger residual NS309-induced relaxation in the presence of UCL 1684. Reduction of blood flow selectively blunts EDHF relaxation in resistance arteries through inhibition of the function of K(Ca) channels. An increase in blood flow leads to a more prominent role of IK1 channels in this relaxation.


Subject(s)
Endothelium, Vascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Mesenteric Arteries/metabolism , Potassium Channels, Calcium-Activated/biosynthesis , Acetylcholine/pharmacology , Animals , Biological Factors/physiology , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Guanylate Cyclase/pharmacology , Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Mesenteric Arteries/drug effects , Muscle Contraction , Muscle Relaxation , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Oximes/pharmacology , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Rats , Rats, Inbred WKY , Receptors, Cytoplasmic and Nuclear/pharmacology , Small-Conductance Calcium-Activated Potassium Channels , Soluble Guanylyl Cyclase , Splanchnic Circulation , Stress, Mechanical
9.
Hypertension ; 46(1): 71-5, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956110

ABSTRACT

A suboptimal fetal environment increases the risk to develop cardiovascular disease in the adult. We reported previously that intrauterine stress in response to reduced uteroplacental blood flow in the pregnant rat limits fetal growth and compromises renal development, leading to an altered renal function in the adult offspring. Here we tested the hypothesis that high dietary sodium intake in rats with impaired renal development attributable to intrauterine stress, results in increased blood pressure, altered renal function, and organ damage. In rats, intrauterine stress was induced by bilateral ligation of the uterine arteries at day 17 of pregnancy. At the age of 12 weeks, the offspring was given high-sodium drinking water (2% sodium chloride). At the age of 16 weeks, rats were instrumented for monitoring of blood pressure and renal function. After intrauterine stress, litter size and birth weight were reduced, whereas hematocrit at birth was increased. Renal blood flow, glomerular filtration rate, and the glomerular filtration fraction were increased significantly after intrauterine stress. High sodium intake did not change renal function and blood pressure in control animals. However, during high sodium intake in intrauterine stress offspring, renal blood flow, glomerular filtration rate, and the filtration fraction were decreased, and blood pressure was increased. In addition, these animals developed severe albuminuria, an important sign of renal dysfunction. Thus, a suboptimal fetal microenvironment, which impairs renal development, results in sodium-dependent hypertension and albuminuria.


Subject(s)
Blood Pressure/drug effects , Fetal Growth Retardation/physiopathology , Sodium, Dietary/administration & dosage , Aging , Albuminuria/chemically induced , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Female , Glomerular Filtration Rate/drug effects , Kidney/physiopathology , Male , Rats , Rats, Wistar , Renal Circulation/drug effects , Sodium, Dietary/pharmacology
10.
J Vasc Res ; 41(3): 277-90, 2004.
Article in English | MEDLINE | ID: mdl-15192268

ABSTRACT

In small arteries, a chronic blood flow reduction leads to inward hypotrophic remodeling, while a chronic blood flow elevation induces outward hypertrophic remodeling. The RhoA/Rho kinase system was shown to be modulated by shear stress, and to be involved in other kinds of vascular remodeling. The aim of this study was to investigate the role of RhoA/Rho kinase in flow-related small artery remodeling. Rat mesenteric small arteries were subjected to flow-modifying surgery. After 1, 2, 4, 16, and 32 days, the animals were sacrificed and small arteries were harvested. Messenger RNA was isolated and amplified. Using cDNA microarray analysis, the differential expression of >14,000 genes was analyzed, part of which was confirmed by RT-PCR. In vivo treatment with fasudil (3 mg/kg/day s.c.) was used to test the effect of Rho kinase inhibition. The main findings are that: (1) blood flow alteration modified the expression of approximately 5% of the genes by >2-fold, (2) flow reduction downregulated many RhoA-related cytoskeletal markers of smooth muscle cell phenotype, (3) many RhoA-related genes were rapidly (<1 day) regulated and (4) fasudil treatment potentiated the inward hypotrophic remodeling in response to chronically reduced flow. These results indicate the importance of the RhoA/Rho kinase system in flow-related small artery remodeling.


Subject(s)
Mesenteric Arteries/physiopathology , Protein Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Male , Mesenteric Arteries/metabolism , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Wistar , Regional Blood Flow , rho-Associated Kinases , rhoA GTP-Binding Protein/genetics
11.
Hypertension ; 43(6): 1283-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15117909

ABSTRACT

Fetal malnutrition and hypoxia may modify organ system maturation and result in cardiovascular diseases in the adult. We tested whether intrauterine stress (IUS) leads to persistent alterations of renal biology. In rats, intrauterine stress was induced by ligation of the uterine arteries at day 17 of pregnancy. Renal arteries of the 21-day-old male offspring were isolated to study pharmacological reactivity. Kidneys were dissected to analyze renal structure and beta-adrenoceptor expression. At 21 days of age, half of the animals underwent unilateral left nephrectomy. At the age of 12 weeks, rats were instrumented for blood pressure monitoring, blood sampling, and renal function measurements. After IUS, litter size and birth weight were reduced, whereas the hematocrit was increased. Renal arterial responses to beta-adrenergic stimulation and sensitivity to adenylyl cyclase activation were increased, along with the renal expression of beta2-adrenoceptors. At 21 days and at 6 months of age, the number and density of the glomeruli were reduced, whereas their size was increased. The filtration fraction and urinary albumin concentration were increased 12 weeks after intrauterine stress. In control rats, removal of the left kidney at 21 days of age did not affect kidney function and blood pressure. However, after IUS, the remaining right kidney failed to compensate for the loss of the left kidney, and blood pressure was increased. In conclusion, prenatal stress transiently modifies renal arterial reactivity and results in long-lasting adverse effects on renal structure and function and on renal compensatory mechanisms.


Subject(s)
Fetal Distress/physiopathology , Kidney Glomerulus/physiopathology , Placenta/blood supply , Renal Artery/physiopathology , Uterus/blood supply , Adaptation, Physiological , Adenylyl Cyclases/metabolism , Adrenergic Agents/pharmacology , Adrenergic Fibers/physiology , Albuminuria/etiology , Animals , Birth Weight , Colforsin/pharmacology , Creatinine/blood , Female , Fetal Distress/etiology , Hematocrit , Hypertension, Renal/etiology , Isoproterenol/pharmacology , Kidney Glomerulus/embryology , Kidney Glomerulus/ultrastructure , Ligation , Litter Size , Male , Nephrectomy , Phenylephrine/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-2/genetics , Renal Artery/drug effects , Renal Artery/embryology , Serotonin/pharmacology , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...