Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Int J Mol Sci ; 21(22)2020 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-33218054

RESUMO

The concept that hypertension and chronic kidney disease (CKD) originate in early life has emerged recently. During pregnancy, tryptophan is crucial for maternal protein synthesis and fetal development. On one hand, impaired tryptophan metabolic pathway in pregnancy impacts fetal programming, resulting in the developmental programming of hypertension and kidney disease in adult offspring. On the other hand, tryptophan-related interventions might serve as reprogramming strategies to prevent a disease from occurring. In the present review, we aim to summarize (1) the three major tryptophan metabolic pathways, (2) the impact of tryptophan metabolism in pregnancy, (3) the interplay occurring between tryptophan metabolites and gut microbiota on the production of uremic toxins, (4) the role of tryptophan-derived metabolites-induced hypertension and CKD of developmental origin, (5) the therapeutic options in pregnancy that could aid in reprogramming adverse effects to protect offspring against hypertension and CKD, and (6) possible mechanisms linking tryptophan metabolism to developmental programming of hypertension and kidney disease.


Assuntos
Desenvolvimento Fetal/fisiologia , Hipertensão/metabolismo , Redes e Vias Metabólicas/fisiologia , Insuficiência Renal Crônica/metabolismo , Triptofano/metabolismo , Animais , Feminino , Microbioma Gastrointestinal/fisiologia , Humanos , Hipertensão/embriologia , Gravidez , Insuficiência Renal Crônica/embriologia
2.
Part Fibre Toxicol ; 16(1): 7, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30691489

RESUMO

BACKGROUND: Exposure to particulate matter (PM) is associated with an adverse intrauterine environment, which can promote adult cardiovascular disease (CVD) risk. Ultrafine particles (UFP) (small size and large surface area/mass ratio) are systemically distributed, induce inflammation and oxidative stress, and have been associated with vascular endothelial dysfunction and arterial vasoconstriction, increasing hypertension risk. Placental stress and alterations in methylation of promoter regions of renin-angiotensin system (RAS)-related elements could be involved in UFP exposure-related programming of hypertension. We investigated whether in utero UFP exposure promotes placental stress by inflammation and oxidative stress, alterations in hydroxysteroid dehydrogenase 11b-type 2 (HSD11B2) and programming of RAS-related elements, and result in altered blood pressure in adult offspring. UFP were collected from ambient air using an aerosol concentrator and physicochemically characterized. Pregnant C57BL/6J pun/pun female mice were exposed to collected UFP (400 µg/kg accumulated dose) by intratracheal instillation and compared to control (nonexposed) and sterile H2O (vehicle) exposed mice. Embryo reabsorption and placental stress by measurement of the uterus, placental and fetal weights, dam serum and fetal cortisol, placental HSD11B2 DNA methylation and protein levels, were evaluated. Polycyclic aromatic hydrocarbon (PAH) biotransformation (CYP1A1 and NQO1 (NAD(P)H dehydrogenase (quinone)1)) enzymes, inflammation and oxidative stress in placentas and fetuses were measured. Postnatal day (PND) 50 in male offspring blood pressure was measured. Methylation and protein expression of (RAS)-related elements, angiotensin II receptor type 1 (AT1R) and angiotensin I-converting enzyme (ACE) in fetuses and lungs of PND 50 male offspring were also assessed. RESULTS: In utero UFP exposure induced placental stress as indicated by an increase in embryo reabsorption, decreases in the uterus, placental, and fetal weights, and HSD11B2 hypermethylation and protein downregulation. In utero UFP exposure induced increases in the PAH-biotransforming enzymes, intrauterine oxidative damage and inflammation and stimulated programming and activation of AT1R and ACE, which resulted in increased blood pressure in the PND 50 male offspring. CONCLUSIONS: In utero UFP exposure promotes placental stress through inflammation and oxidative stress, and programs RAS-related elements that result in altered blood pressure in the offspring. Exposure to UFP during fetal development could influence susceptibility to CVD in adulthood.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/toxicidade , Placenta/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Sistema Renina-Angiotensina/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Feminino , Desenvolvimento Fetal , Hipertensão/induzido quimicamente , Hipertensão/embriologia , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Peptidil Dipeptidase A/metabolismo , Placenta/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Receptor Tipo 1 de Angiotensina/metabolismo , Propriedades de Superfície
4.
Clin Sci (Lond) ; 130(5): 337-48, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26814204

RESUMO

Hypertension is a risk factor for cardiovascular disease, the leading cause of death worldwide. Although multiple factors contribute to the pathogenesis of hypertension, studies by Dr David Barker reporting an inverse relationship between birth weight and blood pressure led to the hypothesis that slow growth during fetal life increased blood pressure and the risk for cardiovascular disease in later life. It is now recognized that growth during infancy and childhood, in addition to exposure to adverse influences during fetal life, contributes to the developmental programming of increased cardiovascular risk. Numerous epidemiological studies support the link between influences during early life and later cardiovascular health; experimental models provide proof of principle and indicate that numerous mechanisms contribute to the developmental origins of chronic disease. Sex has an impact on the severity of cardiovascular risk in experimental models of developmental insult. Yet, few studies examine the influence of sex on blood pressure and cardiovascular health in low-birth weight men and women. Fewer still assess the impact of ageing on sex differences in programmed cardiovascular risk. Thus, the aim of the present review is to highlight current data about sex differences in the developmental programming of blood pressure and cardiovascular disease.


Assuntos
Hipertensão/etiologia , Caracteres Sexuais , Peso ao Nascer/fisiologia , Pressão Sanguínea/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Humanos , Hipertensão/embriologia , Hipertensão/fisiopatologia , Recém-Nascido de Baixo Peso , Recém-Nascido , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Fatores de Risco
5.
J Perinat Med ; 44(6): 705-9, 2016 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26677883

RESUMO

AIM: This study investigates key components of the renin-angiotensin system (RAS) which play a central role in nephrogenesis and possibly in fetal programming of arterial hypertension in adult life. METHODS: We compared a genetic rat model with inborn nephron deficit, the Munich Wistar Fromter rat (MWF), to normotensive Wistar rats during nephrogenesis at day 19 of fetal development (E19) and at postnatal day 7 (D7). RESULTS: At E19 renal mRNA of angiotensin II type 1a (AT1a) (-50%, P<0.05) and type 1b (AT1b) (-55%, P<0.05) receptors were significantly decreased and renal mRNA expression of angiotensin II type 2 (AT2) receptor was fivefold increased in MWF (n=8) as compared to Wistar rats (n=8). At D7 renal mRNA expression of AT1a (-42%, P<0.05) remained lower in MWF (n=8) as compared to Wistar (n=7). Renal mRNA expression of AT2 (-30%, P>0.05) decreased in MWF (n=8) to about the level of the Wistar control (n=6). CONCLUSIONS: Altered fetal expression of key molecules of the renin-angiotensin system in MWF indicates a possible role in genetic low nephron number hypertension.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hipertensão/embriologia , Néfrons/embriologia , Organogênese/fisiologia , Sistema Renina-Angiotensina/genética , Animais , Biomarcadores/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Néfrons/metabolismo , Ratos , Ratos Wistar , Sistema Renina-Angiotensina/fisiologia
6.
Horm Mol Biol Clin Investig ; 18(2): 63-77, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-25390003

RESUMO

BACKGROUND: A substantial body of epidemiological and experimental evidence suggests that a poor fetal and neonatal environment may "program" susceptibility in the offspring to later development of cardiovascular, renal and metabolic diseases. MATERIALS AND METHODS: This review focuses on current knowledge from the available literature regarding the mechanisms linking an adverse developmental environment with an increased risk for cardiovascular, renal and metabolic diseases in adult life. Moreover, this review highlights important sex-dependent differences in the adaptation to developmental insults. RESULTS: Developmental programming of several diseases is secondary to changes in different mechanisms inducing important alterations in the normal development of several organs that lead to significant changes in birth weight. The different diseases occurring as a consequence of an adverse environment during development are secondary to morphological and functional cardiovascular and renal changes, to epigenetic changes and to an activation of several hormonal and regulatory systems, such as angiotensin II, sympathetic activity, nitric oxide, COX2-derived metabolites, oxidative stress and inflammation. The important sex-dependent differences in the developmental programming of diseases seem to be partly secondary to the effects of sex hormones. Recent studies have shown that the progression of these diseases is accelerated during aging in both sexes. CONCLUSIONS: The cardiovascular, renal and metabolic diseases during adult life that occur as a consequence of several insults during fetal and postnatal periods are secondary to multiple structural and functional changes. Future studies are needed in order to prevent the origin and reduce the incidence and consequences of developmental programmed diseases.


Assuntos
Hipertensão/fisiopatologia , Nefropatias/fisiopatologia , Animais , Peso ao Nascer , Epigênese Genética , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Hipertensão/embriologia , Hipertensão/metabolismo , Nefropatias/embriologia , Nefropatias/metabolismo , Masculino , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Estresse Oxidativo , Gravidez , Sistema Renina-Angiotensina/fisiologia , Fatores Sexuais , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Sistema Nervoso Simpático/fisiopatologia
7.
Postepy Hig Med Dosw (Online) ; 68: 899-911, 2014.
Artigo em Polonês | MEDLINE | ID: mdl-24988610

RESUMO

Growth and development in utero is a complex and dynamic process that requires interaction between the mother organism and the fetus. The delivery of macro--and micronutrients, oxygen and endocrine signals has crucial importance for providing a high level of proliferation, growth and differentiation of cells, and a disruption in food intake not only has an influence on the growth of the fetus, but also has negative consequences for the offspring's health in the future. Diseases that traditionally are linked to inappropriate life style of adults, such as type 2 diabetes, obesity, and arterial hypertension, can be "programmed" in the early stage of life and the disturbed growth of the fetus leads to the symptoms of the metabolic syndrome. The structural changes of some organs, such as the brain, pancreas and kidney, modifications of the signaling and metabolic pathways in skeletal muscles and in fatty tissue, epigenetic mechanisms and mitochondrial dysfunction are the basis of the metabolic disruptions. The programming of the metabolic disturbances is connected with the disruption in the intrauterine environment experienced in the early and late gestation period. It causes the changes in deposition of triglycerides, activation of the hormonal "stress axis" and disturbances in the offspring's glucose tolerance. The present review summarizes experimental results that led to the identification of the above-mentioned links and it underlines the role of animal models in the studies of this important concept.


Assuntos
Modelos Animais de Doenças , Doenças Fetais/genética , Doenças Fetais/metabolismo , Doenças Metabólicas/embriologia , Doenças Metabólicas/metabolismo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Anemia/metabolismo , Animais , Encéfalo/embriologia , Diabetes Mellitus Tipo 2/embriologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Gestacional/metabolismo , Epigênese Genética , Feminino , Hipertensão/embriologia , Hipertensão/metabolismo , Rim/embriologia , Doenças Metabólicas/genética , Síndrome Metabólica/embriologia , Síndrome Metabólica/metabolismo , Obesidade/embriologia , Obesidade/metabolismo , Pâncreas/embriologia , Gravidez
8.
Acta Physiol (Oxf) ; 210(2): 307-16, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24268043

RESUMO

Experimental models of developmental programming provide proof of concept and support Barker's original findings that link birthweight and blood pressure. Many experimental models of developmental insult demonstrate a sex difference with male offspring exhibiting a higher blood pressure in young adulthood relative to their age-matched female counterparts. It is well recognized that men exhibit a higher blood pressure relative to age-matched women prior to menopause. Yet, whether this sex difference is noted in individuals born with low birthweight is not clear. Sex differences in the developmental programming of blood pressure may originate from innate sex-specific differences in expression of the renin angiotensin system that occur in response to adverse influences during early life. Sex differences in the developmental programming of blood pressure may also involve the influence of the hormonal milieu on regulatory systems key to the long-term control of blood pressure such as the renin angiotensin system in adulthood. In addition, the sex difference in blood pressure in offspring exposed to a developmental insult may involve innate sex differences in oxidative status or the endothelin system or may be influenced by age-dependent changes in the developmental programming of cardiovascular risk factors such as adiposity. Therefore, this review will highlight findings from different experimental models to provide the current state of knowledge related to the mechanisms that contribute to the aetiology of sex differences in the developmental programming of blood pressure and hypertension.


Assuntos
Desenvolvimento Fetal/fisiologia , Hipertensão/embriologia , Caracteres Sexuais , Animais , Feminino , Humanos , Masculino
9.
Biol Reprod ; 89(6): 144, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24227755

RESUMO

Environmental conditions during perinatal development such as maternal undernutrition, maternal glucocorticoids, placental insufficiency, and maternal sodium overload can program changes in renal Na(+) excretion leading to hypertension. Experimental studies indicate that fetal exposure to an adverse maternal environment may reduce glomerular filtration rate by decreasing the surface area of the glomerular capillaries. Moreover, fetal responses to environmental insults during early life that contribute to the development of hypertension may include increased expression of tubular apical or basolateral membrane Na(+) transporters and increased production of renal superoxide leading to enhanced Na(+) reabsorption. This review will address the role of these potential renal mechanisms in the fetal programming of hypertension in experimental models induced by maternal undernutrition, fetal exposure to glucocorticoids, placental insufficiency, and maternal sodium overload in the rat.


Assuntos
Hipertensão/embriologia , Rim/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Feminino , Taxa de Filtração Glomerular , Glucocorticoides/farmacologia , Humanos , Hipertensão/fisiopatologia , Rim/embriologia , Desnutrição/complicações , Desnutrição/embriologia , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Insuficiência Placentária/fisiopatologia , Gravidez , Ratos , Sódio na Dieta
10.
J Clin Invest ; 123(12): 5052-60, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24270419

RESUMO

Children conceived by assisted reproductive technologies (ART) display a level of vascular dysfunction similar to that seen in children of mothers with preeclamspia. The long-term consequences of ART-associated vascular disorders are unknown and difficult to investigate in healthy children. Here, we found that vasculature from mice generated by ART display endothelial dysfunction and increased stiffness, which translated into arterial hypertension in vivo. Progeny of male ART mice also exhibited vascular dysfunction, suggesting underlying epigenetic modifications. ART mice had altered methylation at the promoter of the gene encoding eNOS in the aorta, which correlated with decreased vascular eNOS expression and NO synthesis. Administration of a deacetylase inhibitor to ART mice normalized vascular gene methylation and function and resulted in progeny without vascular dysfunction. The induction of ART-associated vascular and epigenetic alterations appeared to be related to the embryo environment; these alterations were possibly facilitated by the hormonally stimulated ovulation accompanying ART. Finally, ART mice challenged with a high-fat diet had roughly a 25% shorter life span compared with control animals. This study highlights the potential of ART to induce vascular dysfunction and shorten life span and suggests that epigenetic alterations contribute to these problems.


Assuntos
Anormalidades Cardiovasculares/etiologia , Metilação de DNA , Endotélio Vascular/fisiopatologia , Fertilização in vitro/efeitos adversos , Hipertensão/etiologia , Longevidade , Animais , Aorta/enzimologia , Butiratos/farmacologia , Butiratos/uso terapêutico , Anormalidades Cardiovasculares/embriologia , Dieta Aterogênica , Suscetibilidade a Doenças , Regulação para Baixo , Endotélio Vascular/embriologia , Feminino , Fertilização in vitro/métodos , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Hipertensão/embriologia , Hipertensão/fisiopatologia , Masculino , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/fisiologia , Indução da Ovulação/efeitos adversos , Regiões Promotoras Genéticas , Resistência Vascular/fisiologia , Vasodilatação/fisiologia
11.
J Mol Cell Cardiol ; 65: 108-19, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24140724

RESUMO

During embryonic heart development, the transcription factors Tcf21, Wt1, and Tbx18 regulate activation and differentiation of epicardium-derived cells, including fibroblast lineages. Expression of these epicardial progenitor factors and localization of cardiac fibrosis were examined in mouse models of cardiovascular disease and in human diseased hearts. Following ischemic injury in mice, epicardial fibrosis is apparent in the thickened layer of subepicardial cells that express Wt1, Tbx18, and Tcf21. Perivascular fibrosis with predominant expression of Tcf21, but not Wt1 or Tbx18, occurs in mouse models of pressure overload or hypertensive heart disease, but not following ischemic injury. Areas of interstitial fibrosis in ischemic and hypertensive hearts actively express Tcf21, Wt1, and Tbx18. In all areas of fibrosis, cells that express epicardial progenitor factors are distinct from CD45-positive immune cells. In human diseased hearts, differential expression of Tcf21, Wt1, and Tbx18 also is detected with epicardial, perivascular, and interstitial fibrosis, indicating conservation of reactivated developmental mechanisms in cardiac fibrosis in mice and humans. Together, these data provide evidence for distinct fibrogenic mechanisms that include Tcf21, separate from Wt1 and Tbx18, in different fibroblast populations in response to specific types of cardiac injury.


Assuntos
Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Hipertensão/patologia , Isquemia Miocárdica/patologia , Pericárdio/embriologia , Pericárdio/patologia , Células-Tronco/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Modelos Animais de Doenças , Fibrose Endomiocárdica/embriologia , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Hipertensão/complicações , Hipertensão/embriologia , Hipertensão/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Antígenos Comuns de Leucócito/metabolismo , Leucócitos/metabolismo , Camundongos , Modelos Biológicos , Isquemia Miocárdica/complicações , Isquemia Miocárdica/metabolismo , Pericárdio/metabolismo , Proteínas com Domínio T/metabolismo , Proteínas WT1/metabolismo
13.
Lancet ; 382(9888): 273-83, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23727166

RESUMO

Developmental programming of non-communicable diseases is now an established paradigm. With respect to hypertension and chronic kidney disease, adverse events experienced in utero can affect development of the fetal kidney and reduce final nephron number. Low birthweight and prematurity are the most consistent clinical surrogates for a low nephron number and are associated with increased risk of hypertension, proteinuria, and kidney disease in later life. Rapid weight gain in childhood or adolescence further compounds these risks. Low birthweight, prematurity, and rapid childhood weight gain should alert clinicians to an individual's lifelong risk of hypertension and kidney disease, prompting education to minimise additional risk factors and ensuring follow-up. Birthweight and prematurity are affected substantially by maternal nutrition and health during pregnancy. Optimisation of maternal health and early childhood nutrition could, therefore, attenuate this programming cycle and reduce the global burden of hypertension and kidney disease in the future.


Assuntos
Proteção da Criança , Hipertensão/embriologia , Rim/embriologia , Insuficiência Renal Crônica/embriologia , Adulto , Peso ao Nascer/fisiologia , Pressão Sanguínea/fisiologia , Índice de Massa Corporal , Criança , Desenvolvimento Infantil/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Taxa de Filtração Glomerular/fisiologia , Humanos , Hipertensão/genética , Recém-Nascido de Baixo Peso , Recém-Nascido , Recém-Nascido Prematuro , Rim/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Bem-Estar Materno , Néfrons/patologia , Gravidez , Complicações na Gravidez , Proteinúria/etiologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Fatores de Risco , Aumento de Peso/fisiologia
14.
Ontogenez ; 44(2): 77-90, 2013.
Artigo em Russo | MEDLINE | ID: mdl-23785845

RESUMO

A qualitative and quantitative ultrastructural study of right atrial cardiomyocytes in WAG (normotensive control) and ISIAH (inherited stress-induced arterial hypertension) rats of different age (on day 18 of embryogenesis, on days 12 and 21 after birth, and at an age of 6 and 13 months) was performed. It was shown that, in embryos with an as yet incomplete atrial morphogenesis, secretory granules containing natriuretic peptides are actively formed, accumulated, and dissolved. In postnatal ontogeny, the secretory product is accumulated in atrial cells. In all ontogeny stages studied, the numerical density of secretory granules in the myoendocrine cells of hypertensive rats is greater and the qualitative composition of these granules is more diverse than in the control. It was established that, in atrial myocytes of ISIAH rats, the morphological signs of natriuretic peptide hypersecretion precede the development of genetically programmed high blood pressure. In adult hypertensive rats, hypertrophic and degenerative changes in myocytes are accompanied by excessive accumulation of secretory granules, some of which undergo intracellular degradation.


Assuntos
Envelhecimento/patologia , Átrios do Coração/ultraestrutura , Hipertensão/patologia , Miocárdio/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Animais , Fator Natriurético Atrial/metabolismo , Pressão Sanguínea , Embrião de Mamíferos , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Hipertensão/embriologia , Hipertensão/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Ratos Endogâmicos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura
15.
Ultrasound Obstet Gynecol ; 41(2): 177-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23023990

RESUMO

OBJECTIVES: To examine whether intrauterine growth restriction (IUGR) is associated with increased cardiovascular risk later in life. METHODS: We examined 19 young adults (aged 22-25 years) who were born at term after IUGR, along with 18 controls. All had been examined previously with fetal Doppler, and in the present follow-up with echocardiography, carotid echo-tracking ultrasound, applanation tonometry, blood pressure and laser Doppler, in order to characterize their cardiac and vascular geometry and/or function. RESULTS: The diameter of the ascending aorta and the left ventricular diameter were smaller in the IUGR group, but only ascending aortic diameter remained significantly smaller after adjustment for body surface area (P < 0.05). The aortic pressure augmentation index was higher in the IUGR group (P < 0.05). The common carotid artery diameter, intima-media thickness and distensibility as well as left ventricular mass and function were similar in the two groups. IUGR status was found to be an independent predictor of ascending aortic diameter. CONCLUSIONS: IUGR due to placental dysfunction seems to contribute to the higher systolic blood pressure augmentation and the smaller aortic dimensions that are observed in adults more than 20 years later, with possible negative consequences for future left ventricular performance due to increased aortic impedance.


Assuntos
Retardo do Crescimento Fetal/fisiopatologia , Feto/irrigação sanguínea , Hipertensão/embriologia , Doenças Placentárias/fisiopatologia , Adulto , Aorta/patologia , Pressão Sanguínea/fisiologia , Espessura Intima-Media Carotídea , Feminino , Retardo do Crescimento Fetal/patologia , Seguimentos , Antebraço/irrigação sanguínea , Frequência Cardíaca/fisiologia , Ventrículos do Coração/patologia , Humanos , Hipertensão/patologia , Hipertensão/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Doenças Placentárias/patologia , Gravidez , Fatores de Risco , Adulto Jovem
17.
Hypertension ; 61(1): 180-6, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23150514

RESUMO

Small-for-gestational-age infants are known to develop hypertension in adulthood. This prenatal programming of hypertension (PPH) can result from several insults including maternal dietary protein deprivation, uteroplacental insufficiency, and prenatal administration of glucocorticoids. The mechanisms underlying the development of hypertension remain unclear although the sympathetic nervous system has been indirectly implicated. This study was designed to directly measure renal sympathetic nerve activity both at rest and during physical stress in an animal model of PPH. The adult male offspring of rats fed either a 6% (PPH) or 20% protein diet (control) were investigated. Conscious systolic blood pressure measured by tail cuff was significantly higher in PPH compared with control (140 ± 3 versus 128 ± 3 mm Hg; P<0.05). Baseline mean arterial pressure, heart rate, and renal sympathetic activity were not different between groups during isoflurane anesthesia or after decerebration. Physical stress was induced in decerebrate animals by activating the exercise pressor reflex during static muscle contraction. Stimulation of the exercise pressor reflex evoked significantly larger changes from baseline in mean arterial pressure (40 ± 7 versus 20 ± 4 mm Hg; P<0.05), heart rate (19 ± 3 versus 5 ± 1 bpm; P<0.05), and renal sympathetic activity (198 ± 29% versus 68 ± 14%; P<0.05) in PPH as compared with control. The data demonstrate that the sympathetic response to physical stress is markedly exaggerated in PPH and may play a significant role in the development of hypertension in adults born small for gestational age.


Assuntos
Pressão Sanguínea/fisiologia , Desenvolvimento Fetal/fisiologia , Hipertensão/embriologia , Hipertensão/fisiopatologia , Estresse Fisiológico/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Animais , Feminino , Frequência Cardíaca/fisiologia , Rim/inervação , Rim/fisiopatologia , Masculino , Contração Muscular/fisiologia , Músculo Esquelético/inervação , Músculo Esquelético/fisiopatologia , Gravidez , Ratos , Ratos Sprague-Dawley , Reflexo/fisiologia
18.
PLoS One ; 7(8): e43548, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22927987

RESUMO

OBJECTIVE: Prenatal maternal stress could have permanent effects on the offspring's tissue structure and function, which may predispose to cardiovascular diseases. We investigated whether maternal psychosocial stress is a prenatal factor affecting the blood pressure (BP) of offspring. STUDY DESIGN: In the Amsterdam Born Children and their Development (ABCD) study, around gestational week 16, depressive symptoms, state-anxiety, pregnancy-related anxiety, parenting daily hassles and job strain were recorded by questionnaire. A cumulative stress score was also calculated (based on 80(th) percentiles). Systolic and diastolic BP and mean arterial pressure (MAP) were measured in the offspring at age 5-7 years. Inclusion criteria were: no use of antihypertensive medication during pregnancy; singleton birth; no reported cardiovascular problems in the child (N = 2968 included). RESULTS: After adjustment for confounders, the single stress scales were not associated with systolic and diastolic BP, MAP and hypertension (p>0.05). The presence of 3-4 psychosocial stressors prenatally (4%) was associated with 1.5 mmHg higher systolic and diastolic BP (p = 0.046; p = 0.04) and 1.5 mmHg higher MAP in the offspring (p = 0.02) compared to no stressors (46%). The presence of 3-4 stressors did not significantly increase the risk for hypertension (OR 1.8; 95% CI 0.93.4). Associations did not differ between sexes. Bonferroni correction for multiple testing rendered all associations non-significant. CONCLUSIONS: The presence of multiple psychosocial stressors during pregnancy was associated with higher systolic and diastolic BP and MAP in the child at age 5-7. Further investigation of maternal prenatal stress may be valuable for later life cardiovascular health.


Assuntos
Pressão Sanguínea , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Estresse Psicológico , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Hipertensão/embriologia , Hipertensão/prevenção & controle , Masculino , Gravidez , Caracteres Sexuais
19.
Artigo em Inglês | MEDLINE | ID: mdl-22484708

RESUMO

Effects of dehydration on reptilian embryonic cardiovascular function are unknown. Here, we present the first morphological and physiological data quantifying the cumulative effects of four acute dehydration events on the embryonic American alligator, Alligator mississipiensis. We hypothesized that dehydration would alter embryonic morphology, reduce blood volume and augment the response to angiotensin II (Ang II), a key osmotic and blood volume regulatory response element in adult vertebrates. Drying events at 30%, 40%, 50%, and 60% of embryonic incubation reduced total egg water content by 14.43 ± 0.37 g, a 3.4 fold increase relative to controls. However, embyronic blood volume was greater in the dehydration group at 70% of embryonic incubation compared to controls (0.39 ± 0.044 mLg(-1) and 0.22 ± 0.03 mLg(-1), respectively), however, both groups were similar at 90% of incubation (0.18 ± 0.02 mLg(-1) in the controls and 0.23 ± 0.03 mLg(-1) in the dehydrated group). Dehydration altered the morphological phenotype and resulted in an overall reduction in embryonic mass at both incubation time points measured. Dehydration also altered the physiological phenotype, resulting in embryonic alligators that were relatively bradycardic at 90% of incubation. Arterial Ang II injections resulted in a dose dependent hypertension, which increased in intensity over the span of incubation studied. While progressive incubation altered the Ang II response, dehydration had no impact on the cardiovascular responses to the peptide. Quantification of Ang II type-1 receptor protein using western blot analysis illustrated that dehydration condition and incubation time point did not alter protein quantity. Collectively, our results show that dehydration during embryonic development of the American alligator alters embryonic morphology and baseline heart rate without altering arterial pressure and response to Ang II.


Assuntos
Jacarés e Crocodilos/embriologia , Sistema Cardiovascular/embriologia , Desidratação/fisiopatologia , Jacarés e Crocodilos/metabolismo , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Volume Sanguíneo/efeitos dos fármacos , Volume Sanguíneo/fisiologia , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Desidratação/embriologia , Desidratação/metabolismo , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hipertensão/induzido quimicamente , Hipertensão/embriologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Receptor Tipo 1 de Angiotensina/metabolismo , Zigoto/crescimento & desenvolvimento , Zigoto/metabolismo , Zigoto/fisiologia
20.
Braz. j. med. biol. res ; 44(9): 899-904, Sept. 2011. ilus
Artigo em Inglês | LILACS | ID: lil-599660

RESUMO

Epidemiological and experimental studies have led to the hypothesis of the fetal origin of adult diseases, suggesting that some adult diseases might be determined before birth by altered fetal development. Maternal diabetes subjects the fetus to an adverse environment that has been demonstrated to result in metabolic, cardiovascular and renal impairment in the offspring. The growing amount of obesity in young females in developed and some developing countries should contribute to increasing the incidence of diabetes among pregnant women. In this review, we discuss how renal and extrarenal mechanisms participate in the genesis of hypertension induced by a diabetic status during fetal development.


Assuntos
Animais , Feminino , Humanos , Camundongos , Gravidez , Ratos , Diabetes Mellitus , Hipertensão/embriologia , Gravidez em Diabéticas , Diabetes Mellitus/metabolismo , Rim/metabolismo , Rim/fisiopatologia , Óxido Nítrico/biossíntese , Gravidez em Diabéticas/metabolismo , Fatores de Risco , Sistema Renina-Angiotensina/fisiologia , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...