Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Physiol ; 12: 620058, 2021.
Article in English | MEDLINE | ID: mdl-34603067

ABSTRACT

Fetal growth needs adequate blood perfusion from both sides of the placenta, on the maternal side through the uterine vessels and on the fetal side through the umbilical cord. In a model of intrauterine growth restriction (IUGR) induced by reduced blood volume expansion, uterine artery remodeling was blunted. The aim of this study is to determine if IUGR and fetus sex alter the functional and mechanical parameters of umbilical cord blood vessels. Pregnant rats were given a low sodium (IUGR) or a control diet for the last 7 days of pregnancy. Umbilical arteries and veins from term (22 day) fetal rats were isolated and set-up in wire myographs. Myogenic tone, diameter, length tension curve and contractile response to thromboxane analog U46619 and serotonin (5-HT) were measured. In arteries from IUGR fetuses, myogenic tone was increased in both sexes while diameter was significantly greater only in male fetuses. In umbilical arteries collected from the control group, the maximal contraction to U46619 was lower in females than males. Compared to the control groups, the maximal response decreased in IUGR male arteries and increased in female ones, thus abolishing the sexual dimorphism observed in the control groups. Reduced contractile response to U46619 was observed in the IUGR vein of both sexes. No difference between groups was observed in response to 5HT in arteries. In conclusion, the change in parameters of the umbilical cord blood vessels in response to a mild insult seems to show adaptation that favors better exchange of deoxygenated and wasted blood from the fetus to the placenta with increased myogenic tone.

2.
J Clin Endocrinol Metab ; 106(11): 3295-3311, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34245263

ABSTRACT

CONTEXT: Intrauterine growth restriction (IUGR) is an immediate outcome of an adverse womb environment, exposing newborns to developing cardiometabolic disorders later in life. OBJECTIVE: This study investigates the cardiac metabolic consequences and underlying mechanism of energy expenditure in developing fetuses under conditions of IUGR. METHODS: Using an animal model of IUGR characterized by uteroplacental vascular insufficiency, mitochondrial function, gene profiling, lipidomic analysis, and transcriptional assay were determined in fetal cardiac tissue and cardiomyocytes. RESULTS: IUGR fetuses exhibited an upregulation of key genes associated with fatty acid breakdown and ß-oxidation (Acadvl, Acadl, Acaa2), and mitochondrial carnitine shuttle (Cpt1a, Cpt2), instigating a metabolic gene reprogramming in the heart. Induction of Ech1, Acox1, Acox3, Acsl1, and Pex11a indicated a coordinated interplay with peroxisomal ß-oxidation and biogenesis mainly observed in females, suggesting sexual dimorphism in peroxisomal activation. Concurring with the sex-related changes, mitochondrial respiration rates were stronger in IUGR female fetal cardiomyocytes, accounting for enhanced adenosine 5'-triphosphate production. Mitochondrial biogenesis was induced in fetal hearts with elevated expression of Ppargc1a transcript specifically in IUGR females. Lipidomic analysis identified the accumulation of arachidonic, eicosapentaenoic, and docosapentaenoic polyunsaturated long-chain fatty acids (LCFAs) in IUGR fetal hearts, which leads to nuclear receptor peroxisome proliferator-activated receptor α (PPARα) transcriptional activation in cardiomyocytes. Also, the enrichment of H3K27ac chromatin marks to PPARα-responsive metabolic genes in IUGR fetal hearts outlines an epigenetic control in the early metabolic energy switch. CONCLUSION: This study describes a premature and sex-related remodeling of cardiac metabolism in response to an unfavorable intrauterine environment, with specific LCFAs that may serve as predictive effectors leading to IUGR.


Subject(s)
Energy Metabolism , Fatty Acids/metabolism , Fetal Growth Retardation/pathology , Fetal Heart/pathology , Mitochondria/pathology , Myocytes, Cardiac/pathology , Animals , Animals, Newborn , Female , Fetal Growth Retardation/metabolism , Fetal Heart/metabolism , Male , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Rats, Sprague-Dawley , Sex Factors
3.
Int J Mol Sci ; 19(5)2018 05 21.
Article in English | MEDLINE | ID: mdl-29883404

ABSTRACT

Uncovering the biological role of nuclear receptor peroxisome proliferator-activated receptors (PPARs) has greatly advanced our knowledge of the transcriptional control of glucose and energy metabolism. As such, pharmacological activation of PPARγ has emerged as an efficient approach for treating metabolic disorders with the current use of thiazolidinediones to improve insulin resistance in diabetic patients. The recent identification of growth hormone releasing peptides (GHRP) as potent inducers of PPARγ through activation of the scavenger receptor CD36 has defined a novel alternative to regulate essential aspects of lipid and energy metabolism. Recent advances on the emerging role of CD36 and GHRP hexarelin in regulating PPARγ downstream actions with benefits on atherosclerosis, hepatic cholesterol biosynthesis and fat mitochondrial biogenesis are summarized here. The response of PPARγ coactivator PGC-1 is also discussed in these effects. The identification of the GHRP-CD36-PPARγ pathway in controlling various tissue metabolic functions provides an interesting option for metabolic disorders.


Subject(s)
CD36 Antigens/metabolism , Metabolic Diseases/metabolism , PPAR gamma/metabolism , Signal Transduction , Animals , CD36 Antigens/agonists , Drug Discovery , Energy Metabolism/drug effects , Fatty Acids/metabolism , Humans , Insulin Resistance , Metabolic Diseases/drug therapy , Metabolic Diseases/pathology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , PPAR gamma/agonists , Signal Transduction/drug effects
4.
Am J Physiol Heart Circ Physiol ; 315(2): H415-H422, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29727218

ABSTRACT

Insufficient development of the uteroplacental circulation may contribute to the development of intrauterine growth restriction (IUGR). We developed a rat model of IUGR by administering a low-Na+ diet. This diet reduces maternal blood volume expansion and uteroplacental perfusion. We hypothesized that an impaired endothelial function in radial arteries decreases vasorelaxation and lowers placental perfusion in this IUGR model. The objective was to assess radial uterine artery responses to vasoactive agents in the IUGR model versus controls. The vasoactive agents included phenylephrine and carbachol, use of a pressurized artery myograph, in the absence or presence of inhibitors of nitric oxide (NO) synthase [ N-nitro-l-arginine methyl ester (l-NAME)], cyclooxygenase (Ibuprofen), and endothelium-dependent hyperpolarization {apamin/1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole}, allowing better characterization of the mechanism implicated in endothelium-dependent relaxation. The results show that 1) the diameter of uterine radial arteries was significantly decreased in the IUGR group; 2) sensitivity to phenylephrine was reduced in IUGR arteries, which could be returned to control group values by inhibition of NO production; 3) the relaxation response to carbachol was increased in IUGR rats, principally mediated by endothelium-dependent hyperpolarization in both groups; 4) NO synthase inhibition by l-NAME decreased the maximum relaxation to carbachol only in the IUGR group; and 5) relaxation response to NO donors is increased in IUGR compared with control radial arteries. Contrary to the hypothesis, results in the IUGR model indicate that the NO pathway is activated in radial uterine arteries, most likely in compensation for the reduction in blood uteroplacental perfusion. NEW & NOTEWORTHY In contrast to genetic or surgical models of intrauterine growth restriction, the diet-induced model of reduced maternal volume expansion shows the nitric oxide pathway to be activated in the uterine artery, possibly from increased shear stress and/or placental factors.


Subject(s)
Fetal Growth Retardation/metabolism , Nitric Oxide/metabolism , Uterine Artery/metabolism , Animals , Cyclooxygenase Inhibitors/pharmacology , Female , Ibuprofen/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Uterine Artery/drug effects , Uterine Artery/physiology , Vasodilation
5.
PLoS One ; 11(1): e0145982, 2016.
Article in English | MEDLINE | ID: mdl-26727492

ABSTRACT

Lower maternal plasma volume expansion was found in idiopathic intrauterine growth restriction (IUGR) but the link remains to be elucidated. An animal model of IUGR was developed by giving a low-sodium diet to rats over the last week of gestation. This treatment prevents full expansion of maternal circulating volume and the increase in uterine artery diameter, leading to reduced placental weight compared to normal gestation. We aimed to verify whether this is associated with reduced remodeling of uteroplacental circulation and placental hypoxia. Dams were divided into two groups: IUGR group and normal-fed controls. Blood velocity waveforms in the main uterine artery were obtained by Doppler sonography on days 14, 18 and 21 of pregnancy. On day 22 (term = 23 days), rats were sacrificed and placentas and uterine radial arteries were collected. Diameter and myogenic response of uterine arteries supplying placentas were determined while expression of hypoxia-modulated genes (HIF-1α, VEGFA and VEGFR2), apoptotic enzyme (Caspase -3 and -9) and glycogen cells clusters were measured in control and IUGR term-placentas. In the IUGR group, impaired blood velocity in the main uterine artery along with increased resistance index was observed without alteration in umbilical artery blood velocity. Radial uterine artery diameter was reduced while myogenic response was increased. IUGR placentas displayed increased expression of hypoxia markers without change in the caspases and increased glycogen cells in the junctional zone. The present data suggest that reduced placental and fetal growth in our IUGR model may be mediated, in part, through reduced maternal uteroplacental blood flow and increased placental hypoxia.


Subject(s)
Disease Models, Animal , Fetal Growth Retardation/blood , Placenta/blood supply , Animals , Apoptosis , Biomarkers/blood , Female , Placenta/diagnostic imaging , Placenta/pathology , Pregnancy , Rats , Rats, Sprague-Dawley , Ultrasonography , Umbilical Arteries/physiopathology
6.
Hypertension ; 63(4): 846-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24470461

ABSTRACT

C-Atrial natriuretic peptide (ANP)4-23, a ring deleted analog of ANP that specifically interacts with natriuretic peptide receptor-C (NPR-C), has been shown to decrease the enhanced expression of Giα proteins implicated in the pathogenesis of hypertension. In the present study, we investigated whether in vivo treatment of spontaneously hypertensive rats (SHRs) with C-ANP4-23 could attenuate the development of high blood pressure (BP) and explored the underlying mechanisms responsible for this response. Intraperitoneal injection of C-ANP4-23 at the concentration of 2 or 10 nmol/kg body weight to prehypertensive SHRs attenuated the development of high BP, and at 8 weeks it was decreased by ≈20 and 50 mm Hg, respectively; however, this treatment did not affect BP in Wistar-Kyoto rats. C-ANP4-23 treatment of adult SHRs for 2 weeks also attenuated high BP, heart rate, and restored the impaired vasorelaxation toward control levels. In addition, the enhanced levels of superoxide anion (O2(-)), peroxynitrite, NADPH oxidase activity, and the enhanced expression of Giα proteins, NOX4, p47(phox), nitrotyrosine, and decreased levels of endothelial nitric oxide synthase (eNOS or NOS3) and NO in SHRs were attenuated by C-ANP4-23 treatment; however, the altered levels of NPR-A/NPR-C were not affected by this treatment. In conclusion, these results indicate that NPR-C activation by C-ANP4-23 attenuates the development of high BP in SHRs through the inhibition of enhanced levels of Giα proteins and nitroxidative stress and not through eNOS/cGMP pathway and suggest that NPR-C ligand may have the potential to be used as therapeutic agent in the treatment of cardiovascular complications including hypertension.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Hypertension/prevention & control , Hypertension/physiopathology , Nitric Oxide Synthase/physiology , Oxidative Stress/physiology , Receptors, Atrial Natriuretic Factor/therapeutic use , Animals , Blood Pressure/physiology , Cyclic GMP/physiology , Disease Models, Animal , Heart Rate/physiology , Injections, Intraperitoneal , Male , Nitric Oxide Synthase Type III/physiology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Atrial Natriuretic Factor/administration & dosage , Signal Transduction/physiology , Treatment Outcome
7.
PLoS One ; 7(8): e42622, 2012.
Article in English | MEDLINE | ID: mdl-22880055

ABSTRACT

During development, the risk of developing mesial temporal lobe epilepsy (MTLE) increases when the developing brain is exposed to more than one insult in early life. Early life insults include abnormalities of cortical development, hypoxic-ischemic injury and prolonged febrile seizures. To study epileptogenesis, we have developed a two-hit model of MTLE characterized by two early-life insults: a freeze lesion-induced cortical malformation at post-natal day 1 (P1), and a prolonged hyperthermic seizure (HS) at P10. As early life stressors lead to sexual dimorphism in both acute response and long-term outcome, we hypothesized that our model could lead to gender-based differences in acute stress response and long-term risk of developing MTLE. Male and female pups underwent a freeze-lesion induced cortical microgyrus at P1 and were exposed to HS at P10. Animals were monitored by video-EEG from P90 to P120. Pre and post-procedure plasma corticosterone levels were used to measure stress response at P1 and P10. To confirm the role of sex steroids, androgenized female pups received daily testosterone injections to the mother pre-natally and post-natally for nine days while undergoing both insults. We demonstrated that after both insults females did not develop MTLE while all males did. This correlated with a rise in corticosterone levels at P1 following the lesion in males only. Interestingly, all androgenized females showed a similar rise in corticosterone at P1, and also developed MTLE. Moreover, we found that the cortical lesion significantly decreased the latency to generalized convulsion during hyperthermia at P10 in both genders. The cortical dysplasia volumes at adulthood were also similar between male and female individuals. Our data demonstrate sexual dimorphism in long-term vulnerability to develop epilepsy in the lesion + hyperthermia animal model of MTLE and suggest that the response to early-life stress at P1 contributes significantly to epileptogenesis in a gender-specific manner.


Subject(s)
Disease Susceptibility/pathology , Epilepsy/pathology , Sex Characteristics , Stress, Psychological/pathology , Androgens/metabolism , Animals , Animals, Newborn , Behavior, Animal , Benzoxazines , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Corticosterone/blood , Electroencephalography , Epilepsy/physiopathology , Female , Hyperthermia, Induced , Male , Organ Size , Oxazines , Rats , Rats, Sprague-Dawley , Video Recording
8.
J Endocrinol ; 209(1): 85-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21303825

ABSTRACT

In low sodium-induced intrauterine growth restricted (IUGR) rat, foetal adrenal steroidogenesis as well as the adult renin-angiotensin-aldosterone system (RAAS) is altered. The aim of the present study was to determine the expression of cytochrome P450 aldosterone synthase (P450aldo) and of angiotensin II receptor subtypes 1 (AT(1)R) and 2 (AT(2)R) in adult adrenal glands and whether this expression could be influenced by IUGR and by high-salt intake in a sex-specific manner. After 6 weeks of 0.9% NaCl supplementation, plasma renin activity, P450aldo expression and serum aldosterone levels were decreased in all groups. In males, IUGR induced an increase in AT(1)R, AT(2)R, and P450aldo levels, without changes in morphological appearance of the zona glomerulosa (ZG). By contrast, in females, IUGR had no effect on the expression of AT(1)R, but increased AT(2)R mRNA while decreasing protein expression of AT(2)R and P450aldo. In males, salt intake in IUGR rats reduced both AT(1)R mRNA and protein, while for AT(2)R, mRNA levels decreased whereas protein expression increased. In females, salt intake reduced ZG size in IUGR but had no affect on AT(1)R or AT(2)R expression in either group. These results indicate that, in response to IUGR and subsequently to salt intake, P450aldo, AT(1)R, and AT(2)R levels are differentially expressed in males and females. However, despite these adrenal changes, adult IUGR rats display adequate physiological and adrenal responses to high-salt intake, via RAAS inhibition, thus suggesting that extra-adrenal factors likely compensate for ZG alterations induced by IUGR.


Subject(s)
Adrenal Glands/metabolism , Fetal Growth Retardation/metabolism , Renin-Angiotensin System/physiology , Sodium Chloride, Dietary/metabolism , Adrenal Glands/drug effects , Aldosterone/blood , Analysis of Variance , Animals , Blotting, Western , Female , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Renin/blood , Renin-Angiotensin System/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Sodium Chloride, Dietary/administration & dosage , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Zona Glomerulosa/drug effects , Zona Glomerulosa/metabolism
9.
Am J Physiol Regul Integr Comp Physiol ; 298(4): R899-911, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19923365

ABSTRACT

By feeding a low-sodium diet to dams over the last third of gestation, we have developed an animal model of intrauterine growth restriction (IUGR). Given that fetal adrenal development and maturation occur during late gestation in rats, the aim of this study was to evaluate the expression of proteins and enzymes involved in steroidogenesis and catecholamine synthesis in adrenal glands from IUGR fetuses. A gene microarray was performed to investigate for alteration in the pathways participating in hormone production. Results show that increased aldosterone serum concentrations in IUGR fetuses were associated with higher mRNA adrenal levels of angiotensin II receptor type 1 (AT(1)R) and cytochrome P450 aldosterone synthase in response to decreased serum sodium content. Conversely, reduced serum corticosterone concentrations in these fetuses appear to result from alterations in gene expression involved in cholesterol metabolism, such as the augmented apolipoprotein E levels, and in steroidogenesis, like the decreased levels of cytochrome P45011beta-hydroxylase. Furthermore, increased AT(2)R expression and the presence of hypoxia and oxidative stress may, in turn, explain the higher adrenal mRNA levels of enzymes involved in catecholamine synthesis. Despite this increase, catecholamine adrenal content was reduced in males and was similar in females compared with sex-matched controls, suggesting higher catecholamine secretion. This could be associated with the induction of genes involved in inflammation-related, acute-phase response in IUGR fetuses. All of these alterations could have long-lasting health effects and may, hence, be implicated in the pathogenesis of increased blood pressure and cardiac hypertrophy observed in IUGR adult animals from this model.


Subject(s)
Adrenal Glands/embryology , Fetal Growth Retardation/physiopathology , Uterus/physiology , Adrenal Glands/enzymology , Aldosterone/blood , Animals , Cardiomegaly/etiology , Corticosterone/blood , DNA Primers , Disease Models, Animal , Female , Fetal Blood/physiology , Male , Models, Animal , Pregnancy , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uracil-DNA Glycosidase/genetics
10.
Methods Mol Biol ; 550: 225-37, 2009.
Article in English | MEDLINE | ID: mdl-19495707

ABSTRACT

Fetal programming of adult disease is an area of research that has gained considerable attention. Epidemiological studies suggest that adverse intrauterine environment in fetal life is associated with a higher incidence of hypertension and coronary disease. Several mechanisms could contribute to these diseases and be regulated in a tissue-specific manner. The Na(+)-K(+)-ATPase, a membrane-bound enzyme, maintains the Na(+) and K(+) gradients across the plasma membrane of animal cells and therefore provides a mechanism for cell function regulation. Furthermore, in an in vitro model of cardiac hypertrophy, a decrease in the activity of the tricarboxylic acid (TCA) cycle enzyme, aconitase, was observed. We have shown that in our model of fetal programming, these two enzymes were regulated differently in heart and kidney of adult females.


Subject(s)
Aconitate Hydratase/metabolism , Fetal Development , Kidney/enzymology , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Chitin Synthase/metabolism , Electrophoresis, Polyacrylamide Gel , Fetal Growth Retardation , Models, Animal , Rats
11.
Med Sci (Paris) ; 23(11): 944-9, 2007 Nov.
Article in French | MEDLINE | ID: mdl-18021705

ABSTRACT

Despite widespread accessibility to prenatal care, little is known on the mechanisms initiating early maternal adaptation to pregnancy. Moreover, preeclampsia and intrauterine growth retardation remain the most frequent and serious complications of pregnancy. Recent studies, both in humans and in laboratory animals, have shown that very early events in gestation may be important determinants for the continuation of healthy pregnancy. Certain of these early adaptations appear to be linked to the corpus luteum of pregnancy, as ovarian steroid hormones (especially progesterone) would set the basic hemodynamic conditions, more specifically, generalized vasodilation. This new hemodynamic setup initiates a vicious cycle in which the renin - angiotensin - aldosterone system is activated, together with the resetting of the control of antidiuretic hormone secretion relative to plasma osmolality. This leads to a gradual and substantial increase in plasma volume and a parallel increase in cardiac function (both heart rate and stroke volume) with the goal of maintaining blood pressure in the face of the generalised vasodilation. This includes the creation of a functional arterio-venous shunt represented by the utero-placental circulation. By the end of the first trimester, the decrease in peripheral vascular resistance is marked relative to the increase in cardiac output, resulting in a significant decrease in blood pressure that will be maintained until the third trimester. It is proposed that in preeclampsia, these very early events (vasodilation - increased plasma volume) fail to occur, resulting in an absence of the usual decrease in blood pressure, which is normally seen in the second trimester of pregnancy, and hypertension in the third trimester. Experimental animals, especially the rat, are suitable models to study this early maternal adaptation to pregnancy, since both endocrine and hemodynamic changes appear to be similar to humans.


Subject(s)
Cardiovascular Physiological Phenomena , Pregnancy Complications/physiopathology , Pregnancy/physiology , Female , Fetal Growth Retardation/epidemiology , Humans , Pre-Eclampsia/epidemiology , Pregnancy Complications/epidemiology , Prenatal Care
12.
Am J Physiol Heart Circ Physiol ; 293(6): H3713-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17951367

ABSTRACT

The uterine vasculature plays an important role during pregnancy by providing adequate perfusion of the maternal-fetal interface. To this end, substantial remodeling of the uterine vasculature occurs with consequent changes in responsiveness to contractile agents. The purpose of our study was to characterize the vasorelaxant effects of estrogens on vascular smooth muscles of the rat uterine artery during pregnancy and to evaluate the involvement of estrogen receptors (ESR) and nitric oxide synthases (NOS). To do so, we measured NOS expression in the whole uterine and mesenteric circulatory bed by Western blotting. Vasorelaxant effects of 17beta-estradiol (17beta-E(2)) were assessed on endothelium-denuded uterine arteries with wire myographs in the absence and presence of pharmacological modulators [nitro-L-arginine methyl ester (L-NAME), ICI-182780, tamoxifen]. All experiments were performed on arteries from nonpregnant (NP) and late pregnant (P) rats. In the uterine vasculature of the latter group, NOS3 (endothelial NOS) expression was increased, while NOS1 (neuronal NOS) was reduced compared with NP rats. Expression of the NOS2 (inducible NOS) isoform was undetectable in the two groups. Both 17beta-E(2) and 17alpha-E(2) induced uterine artery relaxation, but the latter evoked lower responses. Endothelium-denuded arteries from NP rats showed larger relaxation with 17beta-E(2) than P rats. This larger relaxation disappeared in the presence of L-NAME. The ESR antagonist ICI-182780 did not affect acute relaxation with 17beta-E(2) and 17alpha-E(2). Moreover, membrane-nonpermeant 17beta-E(2):BSA (estradiol conjugated to bovine serum albumin) did not induce any vasorelaxation. Our results indicate that estrogens exert direct acute vasorelaxant effects in smooth muscles of the rat uterine artery that are mediated by mechanisms independent of ESR activation, but with some stereospecificity. Part of this effect, in NP rats only, is due to nitric oxide produced from muscle NOS1.


Subject(s)
Estradiol/metabolism , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Receptors, Estrogen/metabolism , Uterus/blood supply , Vasodilation , Vasodilator Agents/metabolism , Animals , Arteries/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/antagonists & inhibitors , Tamoxifen/pharmacology , Vasodilation/drug effects
13.
Am J Physiol Regul Integr Comp Physiol ; 293(4): R1657-65, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652367

ABSTRACT

Sodium supplementation given for 1 wk to nonpregnant rats induces changes that are adequate to maintain renal and circulatory homeostasis as well as arterial blood pressure. However, in pregnant rats, proteinuria, fetal growth restriction, and placental oxidative stress are observed. Moreover, the decrease in blood pressure and expansion of circulatory volume, normally associated with pregnancy, are prevented by high-sodium intake. We hypothesized that, in these pregnant rats, a loss of the balance between prooxidation and antioxidation, particularly in kidneys and heart, disturbs the normal course of pregnancy and leads to manifestations such as gestational hypertension. We thus investigated the presence of oxidative/nitrosative stress in heart and kidneys following high-sodium intake in pregnant rats. Markers of this stress [8-isoprostaglandin F(2alpha) (8-iso-PGF(2alpha)) and nitrotyrosine], producer of nitric oxide [nitric oxide synthases (NOSs)], and antioxidants [superoxide dismutase (SOD) and catalase] were measured. Then, molecules (Na(+)-K(+)-ATPase and aconitase) or process [apoptosis (Bax and Bcl-2), inflammation (monocyte chemoattractant protein-1, connective tissue growth factor, and TNF-alpha)] susceptible to free radicals was determined. In kidneys from pregnant rats on 1.8% NaCl-water, NOSs, apoptotic index, and nitrotyrosine expression were increased, whereas Na(+)-K(+)-ATPase mRNA and activity were decreased. In the left cardiac ventricle of these rats, heightened nitrotyrosine, 8-iso-PGF(2alpha), and catalase activity together with reduced endothelial NOS protein expression and SOD and aconitase activities were observed. These findings suggest that oxidative/nitrosative stress in kidney and left cardiac ventricle destabilizes the normal course of pregnancy and could lead to gestational hypertension.


Subject(s)
Heart/drug effects , Hypertension, Pregnancy-Induced/chemically induced , Kidney/drug effects , Sodium/pharmacology , Aconitate Hydratase/metabolism , Animals , Apoptosis/physiology , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Female , Gene Expression Regulation, Enzymologic , Inflammation/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Oxidative Stress , Pregnancy , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/metabolism
14.
Reproduction ; 131(2): 331-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16452726

ABSTRACT

Lowering and increasing sodium intake in pregnant rats evoke opposite changes in renin-angiotensin-aldosterone system (RAAS) activity and are associated with alterations of blood volume expansion. As augmented uterine blood flow during gestation is linked to increased circulatory volume, we wanted to determine if low- and high-sodium intakes affect the mechanical properties and angiotensin II (AngII) responses of the uterine vasculature. Non-pregnant and pregnant rats received a normal sodium (0.22% Na+) diet. On the 15th day of gestation some animals were moved to a low-sodium (0.03%) diet, whereas others were given NaCl supplementation as beverage (saline, 0.9% or 1.8%) for 7 days. All rats were killed after 7 days of treatment (eve of parturition). Uterine arcuate arteries (>100 microm) were set up in wire myographs under a tension equivalent to 50 mmHg transmural pressure. The pregnancy-associated increase in diameter of the uterine arteries was significantly attenuated on the low-sodium diet and 1.8% NaCl supplementation. The arcuate arteries of non-pregnant rats on the low-sodium diet showed markedly increased responses to AngII and phenylephrine (Phe). Pregnancy also resulted in heightened responses to AngII and Phe that were significantly reduced for the former agent in rats on the low-sodium diet. Sodium supplementation of non-pregnant rats did not affect the reactivity of the uterine arteries to AngII, but significantly reduced the effect of Phe (1 micromol/l). High salt also significantly diminished the elevated responses to AngII in the arteries of pregnant animals. It was observed that altered sodium intake affects the mechanical and reactive properties of the uterine arcuate arteries more importantly in pregnant than in non-pregnant rats. Low-salt intake similarly affected the reactivity of the uterine arcuate arteries to AngII and Phe, whereas high-salt intake more specifically affected AngII responses. These results showed that perturbations of sodium intake have major impacts on the structure and functions of the uterine arterial circulation, indicating RAAS involvement in uterine vascular remodeling and function during gestation.


Subject(s)
Angiotensin II/metabolism , Renin-Angiotensin System/physiology , Sodium, Dietary/administration & dosage , Uterus/blood supply , Aldosterone/blood , Angiotensin II/pharmacology , Animals , Arteries/anatomy & histology , Arteries/metabolism , Dose-Response Relationship, Drug , Female , Losartan/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myography , Pregnancy , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Renin/blood , Sodium Chloride/pharmacology , Sodium, Dietary/metabolism
15.
J Physiol ; 565(Pt 1): 197-205, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15774537

ABSTRACT

Epidemiological studies link intra-uterine growth restriction (IUGR) with increased incidence of hypertension and cardiac disease in adulthood. Our rat model of IUGR supports this contention and provides evidence for the programming of susceptibility for hypertension in all offspring. Moreover, in the female offspring only, gross anatomical changes (cardiac ventricle to body ratios) and increased left cardiac ventricular atrial natriuretic peptide (ANP) mRNA levels provide evidence for programming of cardiac disease in this gender. The aim of the current study was to measure changes in cardiac tissue that support remodelling that could be implicated in the initiation of hypertrophy. Adult female rats from our IUGR model and age- and sex-matched controls were killed at 12 weeks of age. Left cardiac ventricles were removed and used for monitoring changes in several key genes, Na+,K+-ATPase beta1 protein expression, cardiomyocyte morphology and contractility as well as citrate synthase and aconitase activities. When compared to controls, female offspring of our IUGR rat model exhibit higher expression (mRNA) of ANP and the atrial isoform of the myosin light chain, lower levels of Na+,K+-ATPase beta1 protein, increased cardiomyocyte depth and volume, increased sarcomere length, diminished cardiomyocyte contractility and lower aconitase activity. Female offspring of our IUGR rat model exhibit changes as adults that are consistent with the onset of cardiac remodelling. The decrease in aconitase activity suggests that oxidative stress may be implicated in this response.


Subject(s)
Cardiomegaly/embryology , Cardiomegaly/physiopathology , Fetal Growth Retardation/physiopathology , Muscle Proteins/metabolism , Myocardial Contraction , Ventricular Dysfunction, Left/embryology , Ventricular Remodeling/physiology , Animals , Cardiomegaly/pathology , Female , Fetal Growth Retardation/complications , Fetal Growth Retardation/pathology , Gene Expression Regulation, Developmental , Organ Size , Rats , Rats, Sprague-Dawley
16.
J Physiol ; 562(Pt 3): 937-50, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15539403

ABSTRACT

We previously reported that sodium restriction during pregnancy reduces plasma volume expansion and promotes intra-uterine growth restriction (IUGR) in rats while it activates the renin-angiotensin-aldosterone system (RAAS). In the present study, we proceeded to determine whether expression of the two angiotensin II (ANGII) receptor subtypes (AT(1) and AT(2)) change in relation to maternal water-electrolyte homeostasis and fetal growth. To this end, pregnant (gestation day 15) and non-pregnant Sprague-Dawley rats were randomly assigned to two groups fed either normal, or Na(+)-restricted diets for 7 days. At the end of the treatment period, plasma aldosterone and renin activity as well as plasma and urine electrolytes were measured. Determinations for AT(1) and AT(2) mRNA and protein were made by RNase protection assay and photoaffinity labelling, respectively, using a number of tissues implicated in volume regulation and fetal growth. In non-pregnant rats, Na(+) restriction decreases Na(+) excretion without altering plasma volume, plasma Na(+) concentration or the expression of AT(1) and AT(2) mRNA or protein in the tissues examined. In normally fed pregnant rats when compared to non-pregnant controls, AT(1) mRNA increases in the hypothalamus as well as pituitary and declines in uterine arteries, while AT(1) protein decreases in the kidney and AT(2) mRNA declines in the adrenal cortex. In pregnant rats, Na(+) restriction induces a decrease in plasma Na(+), an increase in plasma urea, as well as a decline in renal urea and creatinine clearance rates. Protein levels for both AT(1) and AT(2) in the pituitary and AT(2) mRNA in the adrenal cortex are lower in the Na(+)-restricted pregnant group when compared to normally fed pregnant animals. Na(+) restriction also induces a decrease in AT(1) protein in the placenta. In conclusion, these results suggest that pregnancy may increase sensitivity to Na(+) depletion by the tissue-specific modulation of ANGII receptors. Finally, these receptors may be implicated in the IUGR response to low Na(+).


Subject(s)
Body Fluids/metabolism , Fetal Growth Retardation/metabolism , Gonadal Hormones/blood , Receptors, Angiotensin/metabolism , Sodium, Dietary/metabolism , Sodium/deficiency , Adaptation, Physiological , Animals , Female , Fetal Growth Retardation/etiology , Organ Specificity , Pregnancy , Rats , Rats, Sprague-Dawley , Sodium, Dietary/adverse effects , Tissue Distribution
17.
Am J Physiol Heart Circ Physiol ; 287(4): H1848-56, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15205166

ABSTRACT

Gestation is associated with decreased blood pressure and resistance to the effects of vasoconstrictor agents. A recent study showed that pregnant rats, on increased sodium intake, present physiological changes that resemble those observed in preeclampsia. We investigated the effects of sodium supplementation on reactivity and on potassium and Ca(2+) channel activity in blood vessels during gestation. Sodium supplements, 0.9% or 1.8% NaCl as drinking water, were given to nonpregnant and pregnant rats for 7 days (last week of gestation). Reactivity to phenylephrine (PE), KCl, arginine vasopressin (AVP), and tetraethylammonium (TEA) was measured in aortic rings under modulation of potassium and calcium channels. TEA, a nonselective K(+) channel inhibitor, induced concentration-dependent responses in aortic rings from nonpregnant but not in those from pregnant rats. The response to TEA was restored in rings from pregnant rats after preincubation with 10 mmol/l KCl. Sodium supplementation did not affect the response to TEA in the aortas of pregnant animals. After sodium supplementation, maximum responses to PE and AVP were decreased and increased in aortic rings from nonpregnant and pregnant rats, respectively. Cromakalim (an ATP-sensitive K(+) channel activator)-induced inhibition of the responses to the three vasoconstrictors was more striking in aorta from nonpregnant than pregnant rats on regular diet, whereas it produced similar inhibition in tissues from both groups of animals on 0.9% and 1.8% NaCl. NS-1619 (a Ca(2+)-sensitive K(+) activator) elicited heightened effects in the aortas of pregnant animals receiving 0.9% NaCl supplementation. Nifedipine (a Ca(2+) channel blocker) caused greater inhibition of the contractile responses in tissues from nonpregnant rats on regular diet, and its action was increased in pregnant rats on sodium-supplemented diets. These data demonstrate that augmented sodium intake during gestation in the rat is linked with the reversal of gestational-associated resistance to vasopressors and indicate that this is an experimental model showing some features of gestational hypertension.


Subject(s)
Blood Pressure/physiology , Calcium/metabolism , Potassium/metabolism , Pre-Eclampsia/metabolism , Sodium Chloride, Dietary/pharmacology , Animals , Aorta/physiology , Arginine Vasopressin/pharmacology , Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Female , Male , Nifedipine/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Chloride/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Tetraethylammonium/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
18.
Am J Physiol Heart Circ Physiol ; 285(1): H375-83, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12793982

ABSTRACT

Despite an increase of circulatory volume and of renin-angiotensin-aldosterone system (RAAS) activity, pregnancy is paradoxically accompanied by a decrease in blood pressure. We have reported that the decrease in blood pressure was maintained in pregnant rats despite overactivation of RAAS following reduction in sodium intake. The purpose of this study was to evaluate the impact of the opposite condition, e.g., decreased activation of RAAS during pregnancy in the rat. To do so, 0.9% or 1.8% NaCl in drinking water was given to nonpregnant and pregnant Sprague-Dawley rats for 7 days (last week of gestation). Increased sodium intakes (between 10- and 20-fold) produced reduction of plasma renin activity and aldosterone in both nonpregnant and pregnant rats. Systolic blood pressure was not affected in nonpregnant rats. However, in pregnant rats, 0.9% sodium supplement prevented the decreased blood pressure. Moreover, an increase of systolic blood pressure was obtained in pregnant rats receiving 1.8% NaCl. The 0.9% sodium supplement did not affect plasma and fetal parameters. However, 1.8% NaCl supplement has larger effects during gestation as shown by increased plasma sodium concentration, hematocrit level, negative water balance, proteinuria, and intrauterine growth restriction. With both sodium supplements, decreased AT1 mRNA levels in the kidney and in the placenta were observed. Our results showed that a high-sodium intake prevents the pregnancy-induced decrease of blood pressure in rats. Nonpregnant rats were able to maintain homeostasis but not the pregnant ones in response to sodium load. Furthermore, pregnant rats on a high-sodium intake (1.8% NaCl) showed some physiological responses that resemble manifestations observed in preeclampsia.


Subject(s)
Blood Pressure/physiology , Pregnancy, Animal/physiology , Sodium, Dietary/pharmacology , Animals , Blood Pressure/drug effects , Creatinine/blood , Eating/physiology , Electrolytes/blood , Female , Fetal Weight/drug effects , Fetal Weight/physiology , Hematocrit , Homeostasis/physiology , Hormones/blood , Kidney/drug effects , Kidney/metabolism , Nuclease Protection Assays , Organ Size/drug effects , Organ Size/physiology , Placenta/drug effects , Placenta/physiology , Pregnancy , RNA Probes , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/biosynthesis , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Sodium/blood , Urodynamics/drug effects
19.
Endocrinology ; 143(9): 3333-40, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193545

ABSTRACT

Hormone-sensitive lipase (HSL, E.C.3.1.1.3, gene designation Lipe) is reportedly the major cholesteryl esterase of adrenal cortex. Because of the potential importance of cholesteryl ester hydrolysis in steroidogenesis, gene-targeted HSL-deficient mice were assessed for adrenal cortical morphology and function. Compared with control animals, HSL deficiency results in a marked accumulation of lipid droplets both in zona glomerulosa and zona fasciculata. In the zona fasciculata, lipid accumulation was observed progressively from the outer to the inner regions, culminating near the corticomedullary junction with the formation of syncytial-lipoid structures having the appearance of degenerative cells. These morphological changes did not significantly alter the basal levels of circulating corticosterone, but following ACTH stimulation, corticosterone levels were decreased (P < 0.001). The observation of normal basal corticosterone and aldosterone levels demonstrates that some free cholesterol for steroid synthesis can be produced independently of HSL. Taken together, these results indicate that HSL-deficient mice accumulate lipid droplets in such a way as to impair acute ACTH stimulation of corticosterone secretion. Such observations are also found in some forms of congenital adrenal hyperplasia. By extension, HSL deficiency may be a cause of hereditary adrenocortical hypofunction in humans.


Subject(s)
Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Corticosterone/pharmacology , Lipid Metabolism , Sterol Esterase/deficiency , Adrenal Cortex/drug effects , Adrenal Cortex/ultrastructure , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Medulla/metabolism , Aldosterone/blood , Animals , Cholesterol Esters/metabolism , Corticosterone/blood , Gene Targeting , Hydrolysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondria/ultrastructure , Zona Fasciculata/metabolism , Zona Fasciculata/ultrastructure , Zona Glomerulosa/metabolism , Zona Glomerulosa/ultrastructure
20.
Am J Physiol Endocrinol Metab ; 283(1): E124-31, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12067852

ABSTRACT

Epidemiological studies have produced evidence that unfavorable intrauterine environments during fetal life may lead to adverse outcomes in adulthood. We have previously shown that a low-sodium diet, given to pregnant rats over the last week of gestation, results in intrauterine growth restriction (IUGR). We hypothesize that pups born with IUGR are more susceptible to the development of hypertension in adulthood. IUGR fetuses and rats aged 1 wk were characterized for organ growth and renal morphogenesis. The adults (12 wk) were evaluated for weight, systolic blood pressure, activity of the renin-angiotensin-aldosterone system (RAAS), and renal function; hearts and kidneys underwent a histological examination. Brain and cardiac ventricle-to-body ratios were increased in IUGR fetuses compared with age-matched controls, whereas the kidney-to-body ratio was unchanged. Systolic blood pressure was elevated in both IUGR male and female adults. Plasma aldosterone levels were not correlated with increased plasma renin activity. Moreover, urinary sodium was decreased, whereas plasma urea was elevated in both males and females, and creatinine levels were augmented only in females, suggesting a glomerular filtration impairment in IUGR. In our model of IUGR induced by a low-sodium diet given to pregnant rats, high blood pressure, alteration of the RAAS, and renal dysfunction are observed in adult life. Differences observed between male and female adults suggest the importance of gender in outcomes in adulthood after IUGR.


Subject(s)
Aging , Fetal Growth Retardation/physiopathology , Hypertension/physiopathology , Kidney Diseases/physiopathology , Angiotensins/blood , Animals , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/genetics , Blood Pressure , Body Weight , Corticosterone/blood , Female , Fetal Growth Retardation/complications , Heart Ventricles/cytology , Heart Ventricles/metabolism , Hypertension/complications , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/complications , Kidney Diseases/pathology , Kidney Function Tests , Kidney Glomerulus/pathology , Male , Organ Size , Pregnancy , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Renin/blood , Renin-Angiotensin System
SELECTION OF CITATIONS
SEARCH DETAIL
...