Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Reprod Toxicol ; 99: 168-176, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33049332

ABSTRACT

Intrauterine growth restriction (IUGR) affects 10-15% of all pregnancies worldwide. IUGR may result from maternal, placental or fetal origin. Maternal malnutrition before and during pregnancy represents the most prevalent non-genetic or placental cause. IUGR reflects an abnormal adaptive fetal growth in a deleterious environment. Individuals born after IUGR are more susceptible to develop diseases related to subsequent stressors through a lifetime. Animal models help to decipher the underlying causes of dysregulated pathways and molecular modifications conditioning health and disease in adult offspring born after IUGR. The aim of this review is to summarize current knowledge on long term consequences of IUGR, integrating animal models and human studies for a better care of IUGR-born individuals in a life course perspective.


Subject(s)
Fetal Growth Retardation , Adult , Animals , Disease Susceptibility , Environmental Pollutants/toxicity , Female , Humans , Pregnancy
2.
J Dev Orig Health Dis ; 9(6): 598-614, 2018 12.
Article in English | MEDLINE | ID: mdl-30078383

ABSTRACT

Intrauterine growth restriction (IUGR) has been identified as a risk factor for adult chronic kidney disease (CKD), including hypertension (HTN). Accelerated postnatal catch-up growth superimposed to IUGR has been shown to further increase the risk of CKD and HTN. Although the impact of excessive postnatal growth without previous IUGR is less clear, excessive postnatal overfeeding in experimental animals shows a strong impact on the risk of CKD and HTN in adulthood. On the other hand, food restriction in the postnatal period seems to have a protective effect on CKD programming. All these effects are mediated at least partially by the activation of the renin-angiotensin system, leptin and neuropeptide Y (NPY) signaling and profibrotic pathways. Early nutrition, especially in the postnatal period has a significant impact on the risk of CKD and HTN at adulthood and should receive specific attention in the prevention of CKD and HTN.


Subject(s)
Fetal Growth Retardation/physiopathology , Hypertension/prevention & control , Infant Nutritional Physiological Phenomena/physiology , Nutritional Status/physiology , Renal Insufficiency, Chronic/prevention & control , Animals , Child Development/physiology , Disease Models, Animal , Humans , Hypertension/epidemiology , Hypertension/etiology , Hypertension/physiopathology , Infant, Low Birth Weight/physiology , Infant, Newborn , Leptin/metabolism , Metabolic Networks and Pathways/physiology , Neuropeptide Y/metabolism , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/physiopathology , Renin-Angiotensin System/physiology
3.
J Dev Orig Health Dis ; 9(6): 615-631, 2018 12.
Article in English | MEDLINE | ID: mdl-29909803

ABSTRACT

Epidemiological and experimental observations tend to prove that environment, lifestyle or nutritional challenges influence heart functions together with genetic factors. Furthermore, when occurring during sensitive windows of heart development, these environmental challenges can induce an 'altered programming' of heart development and shape the future heart disease risk. In the etiology of heart diseases driven by environmental challenges, epigenetics has been highlighted as an underlying mechanism, constituting a bridge between environment and heart health. In particular, micro-RNAs which are involved in each step of heart development and functions seem to play a crucial role in the unfavorable programming of heart diseases. This review describes the latest advances in micro-RNA research in heart diseases driven by early exposure to challenges and discusses the use of micro-RNAs as potential targets in the reversal of the pathophysiology.


Subject(s)
Fetal Development/genetics , Heart Diseases/etiology , Heart/embryology , MicroRNAs/physiology , Prenatal Exposure Delayed Effects/etiology , Environmental Exposure/adverse effects , Epigenesis, Genetic/physiology , Female , Gene Expression Regulation, Developmental/physiology , Heart/growth & development , Heart Diseases/prevention & control , Humans , Maternal Exposure/adverse effects , Maternal Nutritional Physiological Phenomena/physiology , Pregnancy , Prenatal Exposure Delayed Effects/prevention & control
4.
Nutr Metab Cardiovasc Dis ; 28(9): 944-951, 2018 09.
Article in English | MEDLINE | ID: mdl-29752038

ABSTRACT

BACKGROUND AND AIMS: The prevalence of obesity is increasing worldwide at an alarming rate. Altered early nutrition, in particular postnatal overfeeding (PNOF), is a risk factor for impaired cardiac function in adulthood. In the understanding of the initiation or progression of heart diseases, NLRP3 inflammasome and non-coding RNAs have been proposed as key players. In this context, the aim of this study was to decipher the role of NLRP3 inflammasome and its post transcriptional control by micro-RNAs in the regulation of cardiac metabolic function induced by PNOF in mice. METHODS AND RESULTS: Based on a model of mice exposed to PNOF through litter size reduction, we observed increased cardiac protein expression levels of NLRP3 and ETS-1 associated with alterations in insulin signaling. Additionally, miR-193b levels were down-regulated in the adult hearts of overfed animals. In a cardiomyocyte cell line, transfection with miR-193b induced down-regulation of ETS-1 and NLRP3 and improved insulin signaling. CONCLUSIONS: These findings suggest that the miR-193b could be involved in cardiac phenotypic changes observed in adulthood induced by PNOF likely through the regulation of ETS-1 and NLRP3 expression, and through this of insulin signaling.


Subject(s)
Animal Nutritional Physiological Phenomena , Heart Diseases/etiology , Inflammasomes/metabolism , Myocardium/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nutritional Status , Overnutrition/complications , Animals , Animals, Newborn , Cell Line , Disease Models, Animal , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/physiopathology , Insulin/metabolism , Litter Size , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Overnutrition/genetics , Overnutrition/metabolism , Overnutrition/physiopathology , Proto-Oncogene Protein c-ets-1/metabolism , Rats , Signal Transduction , Time Factors
5.
J Dev Orig Health Dis ; 8(4): 448-464, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28460648

ABSTRACT

Individuals born after intrauterine growth restriction (IUGR) have an increased risk of perinatal morbidity/mortality, and those who survive face long-term consequences such as cardiovascular-related diseases, including systemic hypertension, atherosclerosis, coronary heart disease and chronic kidney disease. In addition to the demonstrated long-term effects of decreased nephron endowment and hyperactivity of the hypothalamic-pituitary-adrenal axis, individuals born after IUGR also exhibit early alterations in vascular structure and function, which have been identified as key factors of the development of cardiovascular-related diseases. The endothelium plays a major role in maintaining vascular function and homeostasis. Therefore, it is not surprising that impaired endothelial function can lead to the long-term development of vascular-related diseases. Endothelial dysfunction, particularly impaired endothelium-dependent vasodilation and vascular remodeling, involves decreased nitric oxide (NO) bioavailability, impaired endothelial NO synthase functionality, increased oxidative stress, endothelial progenitor cells dysfunction and accelerated vascular senescence. Preventive approaches such as breastfeeding, supplementation with folate, vitamins, antioxidants, L-citrulline, L-arginine and treatment with NO modulators represent promising strategies for improving endothelial function, mitigating long-term outcomes and possibly preventing IUGR of vascular origin. Moreover, the identification of early biomarkers of endothelial dysfunction, especially epigenetic biomarkers, could allow early screening and follow-up of individuals at risk of developing cardiovascular and renal diseases, thus contributing to the development of preventive and therapeutic strategies to avert the long-term effects of endothelial dysfunction in infants born after IUGR.


Subject(s)
Cardiovascular Diseases/physiopathology , Endothelium, Vascular/physiopathology , Fetal Growth Retardation/physiopathology , Kidney Diseases/physiopathology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/epidemiology , Humans , Infant, Newborn , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Nitric Oxide/physiology , Oxidative Stress/physiology , Vasodilation/physiology
6.
Arch Pediatr ; 22(10): 1047-55, 2015 Oct.
Article in French | MEDLINE | ID: mdl-26143998

ABSTRACT

The survival of preterm babies has increased over the last few decades. However, disorders associated with preterm birth, known as oxygen radical diseases of neonatology, such as retinopathy, bronchopulmonary dysplasia, periventricular leukomalacia, and necrotizing enterocolitis are severe complications related to oxidative stress, which can be defined by an imbalance between oxidative reactive species production and antioxidant defenses. Oxidative stress causes lipid, protein, and DNA damage. Preterm infants have decreased antioxidant defenses in response to oxidative challenges, because the physiologic increase of antioxidant capacity occurs at the end of gestation in preparation for the transition to extrauterine life. Therefore, preterm infants are more sensitive to neonatal oxidative stress, notably when supplemental oxygen is being delivered. Furthermore, despite recent advances in the management of neonatal respiratory distress syndrome, controversies persist concerning the oxygenation saturation targets that should be used in caring for preterm babies. Identification of adequate biomarkers of oxidative stress in preterm infants such as 8-iso-prostaglandin F2α, and adduction of malondialdehyde to hemoglobin is important to promote specific therapeutic approaches. At present, no therapeutic strategy has been validated as prevention or treatment against oxidative stress. Breastfeeding should be considered as the main measure to improve the antioxidant status of preterm infants. In the last few years, melatonin has emerged as a protective molecule against oxidative stress, with antioxidant and free-radical scavenger roles, in experimental and preliminary human studies, giving hope that it can be used in preterm infants in the near future.


Subject(s)
Infant, Premature , Oxidative Stress , Advanced Oxidation Protein Products/metabolism , Aldehydes/metabolism , Antioxidants/therapeutic use , Biomarkers/metabolism , Breast Feeding , Delivery Rooms , Female , Humans , Infant, Newborn , Infant, Premature/metabolism , Isoprostanes/metabolism , Malondialdehyde/metabolism , Melatonin/therapeutic use , Oxygen Inhalation Therapy/adverse effects , Parenteral Nutrition/adverse effects , Pregnancy , Reactive Oxygen Species/metabolism , Respiratory Distress Syndrome, Newborn/therapy , Retinopathy of Prematurity/etiology
7.
Best Pract Res Clin Obstet Gynaecol ; 29(2): 256-69, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25267399

ABSTRACT

In the 1980s, David Barker and Colleagues proposed that the major causes of cardiovascular and metabolic diseases have their roots in early development. There is now robust evidence that an hyperglycemic intrauterine environment is responsible not only for significant short-term morbidity in the fetus and the neonate but also for an increased risk of developing diabetes as well as other chronic, noncommunicable diseases at adulthood. The risk is higher in pregestational diabetes, but unrecognized and/or poorly managed gestational diabetes (GDM) may have similar consequences. Although a relatively clear picture of the pathogenesis of the fetal and neonatal complications of maternal diabetes and of their interrelationship is available today, the intimate molecular mechanisms involved in the long term are far from being understood. While the rate of GDM is sharply increasing in association with the pandemic of obesity and of type 2 diabetes over the world, we review here the current understanding of short- and long-term outcomes of fetuses exposed to a diabetic environment.


Subject(s)
Diabetes, Gestational , Pregnancy in Diabetics , Prenatal Exposure Delayed Effects/etiology , Cardiovascular Diseases/etiology , Developed Countries , Diabetes Mellitus, Type 2/etiology , Female , Health Status Disparities , Humans , Infant, Newborn , Infant, Newborn, Diseases/etiology , Kidney Diseases/etiology , Obesity/etiology , Pregnancy , Risk Factors
8.
J Dev Orig Health Dis ; 2(3): 184-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-25141044

ABSTRACT

We have previously shown that neonatal high oxygen (O2) exposure in rats leads to hypertension and vascular dysfunction in adulthood. Pulse-wave velocity (PWV), an indirect measure of vascular biophysical properties (arterial stiffness or distensibility), is a sensitive marker of cardiovascular health. Its measurement in rats is mostly based on invasive hemodynamics measurements, prohibiting longitudinal studies particularly relevant in models of developmental programming of cardiovascular dysfunctions. With this study, we sought (1) to verify the feasibility and validity of measuring of aortic PWV in Sprague-Dawley rats by ultrasound; (2) to use the technique to compare aortic PWV in adult rats exposed to O2 as newborns (80% day 3-10 of life) v. controls; and (3) to develop an algorithm to calculate PWV in a non-invasive manner. We calculated aortic PWV using standard echocardiography and electrocardiogram, and validated the measures with PWV obtained by intraaortic catheters. Aortic full length was measured at sacrifice. PWV was significantly increased in O2 exposed (505 ± 18 cm/s) v. control animals (421 ± 17 cm/s, P < 0.01). With regard to weight, femur length and distance from the manubrium to the anal margin (MA length), the latter showed the best correlation (R = 0.84, P < 0.0001) with full aorta length derived from (L) = 0.339 × (MA length) + 4.281. The current data using echo-Doppler method demonstrated increased aortic stiffness in adult rats exposed to hyperoxia as newborns and suggests that non-invasive longitudinal studies of aortic PWV can be performed using the proposed algorithm for estimation of the full aorta length.

9.
Am J Physiol Regul Integr Comp Physiol ; 291(4): R1060-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16741139

ABSTRACT

The renin-angiotensin system plays a key role in the initiation and maintenance of elevated blood pressure associated with altered intrauterine milieu. The current studies were undertaken to verify whether vascular response to ANG II is increased in adult offspring of low-protein fed dams (LP) compared with control (CTRL) and if so, to examine underlying mechanism(s). ANG II-induced contraction of carotid rings was increased in LP (E(max), the maximum asymptote of the curve, relative to maximal response to KCl 80 mM: 230 +/- 3% LP vs. 201 +/- 2% CTRL, P < 0.05). In both groups, contraction to ANG II was mediated solely by AT1R. Responses to thromboxane A2 analog U-46619 and to KCl 80 mM under step increases in tension were similar between groups. Endothelium depletion enhanced contraction to ANG II in both groups, more so in LP. Blockade of endothelin formation had no effect on response to ANG II, and ANG-(1-7) did not elicit vasomotor response in either group. Superoxide dismutase (SOD) analog Tempol normalized LP without modifying CTRL response to ANG II. Basal levels of superoxide (aortic segments, lucigenin-enhanced chemiluminescence and fluorescent dye hydroethidine) were higher in LP. ANG II further increased superoxide production in LP only, and this was inhibited by coincubation with diphenylene iodonium or apocynin (inhibitor of NADPH oxidase complex). AT1R expression in carotid arteries was increased in LP, whereas SOD expression was unchanged. In conclusion, vasoconstriction to ANG II is exaggerated in this model of developmental programming of hypertension, secondary to enhanced vascular production of superoxide anion by NADPH oxidase with concomitant increase of AT1R expression.


Subject(s)
Angiotensin II/pharmacology , Dietary Proteins/pharmacology , Hypertension/physiopathology , Prenatal Exposure Delayed Effects , Vasoconstrictor Agents/pharmacology , Age Factors , Animal Feed , Animals , Antioxidants/pharmacology , Blotting, Western , Cyclic N-Oxides/pharmacology , Diet, Protein-Restricted , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Male , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Spin Labels , Superoxide Dismutase/metabolism , Vasoconstriction/drug effects , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL