Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Life Sci ; 285: 120016, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34614415

RESUMEN

PURPOSE: Ten percent of pregnancies are affected by intrauterine growth restriction (IUGR), and evidence suggests that affected neonates have reduced activity of hepatic cytochrome P450 (CYP) drug metabolising enzymes. Given that almost all pregnant individuals take medications and additional medications are often required during an IUGR pregnancy, we aimed to determine the impact of IUGR on hepatic CYP activity in sheep fetuses and pregnant ewes. METHODS: Specific probes were used to determine the impact of IUGR on the activity of several CYP isoenzymes (CYP1A2, CYP2C19, CYP2D6 and CYP3A) in sheep fetuses and pregnant ewes. Probes were administered intravenously to the ewe at 132 days (d) gestation (term 150 d), followed by blood sampling from the maternal and fetal circulation over 24 h. Maternal and fetal liver tissue was collected at 139-140 d gestation, from which microsomes were isolated and incubated with probes. Metabolite and maternal plasma cortisol concentrations were measured using Liquid Chromatography - tandem mass spectrometry (LC-MS/MS). RESULTS: Maternal plasma cortisol concentration and maternal hepatic CYP1A2 and CYP3A activity was significantly higher in IUGR pregnancies. Maternal hepatic CYP activity was higher than fetal hepatic CYP activity for all CYPs tested, and there was minimal CYP1A2 or CYP3A activity in the late gestation fetus when assessed using in vitro methods. CONCLUSIONS: The physiological changes to the maternal-placental-fetal unit in an IUGR pregnancy have significant effects on maternal drug metabolism, suggesting changes in medications and/or doses may be required to optimise maternal and fetal health.


Asunto(s)
Corticosterona/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Retardo del Crecimiento Fetal/enzimología , Hidrocortisona/metabolismo , Hígado/enzimología , Intercambio Materno-Fetal , Placenta/metabolismo , Animales , Transporte Biológico , Corticosterona/sangre , Femenino , Hidrocortisona/sangre , Embarazo , Ovinos
2.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R833-R843, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34668428

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a membrane-bound protein containing 805 amino acids. ACE2 shows approximately 42% sequence similarity to somatic ACE but has different biochemical activities. The key role of ACE2 is to catalyze the vasoconstrictor peptide angiotensin (ANG) II to Ang-(1-7), thus regulating the two major counterbalancing pathways of the renin-angiotensin system (RAS). In this way, ACE2 plays a protective role in end-organ damage by protecting tissues from the proinflammatory actions of ANG II. The circulating RAS is activated in normal pregnancy and is essential for maintaining fluid and electrolyte homeostasis and blood pressure. Renin-angiotensin systems are also found in the conceptus. In this review, we summarize the current knowledge on the regulation and function of circulating and uteroplacental ACE2 in uncomplicated and complicated pregnancies, including those affected by preeclampsia and fetal growth restriction. Since ACE2 is the receptor for SARS-CoV-2, and COVID-19 in pregnancy is associated with more severe disease and increased risk of abnormal pregnancy outcomes, we also discuss the role of ACE2 in mediating some of these adverse consequences. We propose that dysregulation of ACE2 plays a critical role in the development of preeclampsia, fetal growth restriction, and COVID-19-associated pregnancy pathologies and suggest that human recombinant soluble ACE2 could be a novel therapeutic to treat and/or prevent these pregnancy complications.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Placenta/enzimología , Complicaciones del Embarazo/enzimología , Sistema Renina-Angiotensina , Útero/enzimología , Enzima Convertidora de Angiotensina 2/uso terapéutico , Animales , Presión Sanguínea , COVID-19/enzimología , COVID-19/fisiopatología , COVID-19/virología , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/fisiopatología , Humanos , Mediadores de Inflamación/metabolismo , Placenta/fisiopatología , Preeclampsia/enzimología , Preeclampsia/fisiopatología , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/fisiopatología , Complicaciones Infecciosas del Embarazo/enzimología , Complicaciones Infecciosas del Embarazo/fisiopatología , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/patogenicidad , Útero/fisiopatología , Equilibrio Hidroelectrolítico
3.
Am J Physiol Regul Integr Comp Physiol ; 320(5): R653-R662, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33621475

RESUMEN

Currently, there is no effective treatment for placental dysfunction in utero. In a ligated mouse model of fetal growth restriction (FGR), nanoparticle-mediated human insulin-like 1 growth factor (hIGF1) gene delivery (NP-Plac1-hIGF1) increased hIGF1 expression and maintained fetal growth. However, whether it can restore fetal growth remains to be determined. Using the endothelial nitric oxide synthase knockout (eNOS-/-) mouse model, a genetic model of FGR, we found that despite inducing expression of hIGF1 in the placentas treated with NP-Plac1-hIGF1 (P = 0.0425), FGR did not resolve. This was associated with no change to the number of fetal capillaries in the placental labyrinth; an outcome which was increased with NP-Plac1-hIGF1 treatment in the ligated mouse model, despite increased expression of angiopoietin 1 (P = 0.05), and suggested IGF1 signaling in the placenta requires eNOS to modulate placenta angiogenesis. To further assess this hypothesis, BeWo choriocarcinoma cell line and human placental explant cultures were treated with NP-Plac1-hIGF1, oxidative stress was induced with hydrogen peroxide (H2O2), and NOS activity was inhibited using the inhibitor NG-monomethyl-l-arginine (l-NMMA). In both BeWo cells and explants, the protective effect of NP-Plac1-hIGF1 treatment against H2O2-induced cell death/lactate dehydrogenase release was prevented by eNOS inhibition (P = 0.003 and P < 0.0001, respectively). This was associated with an increase in mRNA expression of oxidative stress markers hypoxia inducing factor 1α (HIF1α; P < 0.0001) and ADAM10 (P = 0.0002) in the NP-Plac1-hIGF1 + H2O2 + l-NMMA-treated BeWo cells. These findings show for the first time the requirement of eNOS/NOS in IGF1 signaling in placenta cells that may have implications for placental angiogenesis and fetal growth.


Asunto(s)
Retardo del Crecimiento Fetal/terapia , Feto/irrigación sanguínea , Terapia Genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Placenta/irrigación sanguínea , Trofoblastos/enzimología , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Técnicas de Transferencia de Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas , Óxido Nítrico Sintasa de Tipo III/genética , Estrés Oxidativo , Embarazo , Transducción de Señal , Técnicas de Cultivo de Tejidos , Trofoblastos/patología
4.
Am J Physiol Endocrinol Metab ; 319(1): E67-E80, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32396498

RESUMEN

Fetal sheep with placental insufficiency-induced intrauterine growth restriction (IUGR) have lower hindlimb oxygen consumption rates (OCRs), indicating depressed mitochondrial oxidative phosphorylation capacity in their skeletal muscle. We hypothesized that OCRs are lower in skeletal muscle mitochondria from IUGR fetuses, due to reduced electron transport chain (ETC) activity and lower abundances of tricarboxylic acid (TCA) cycle enzymes. IUGR sheep fetuses (n = 12) were created with mid-gestation maternal hyperthermia and compared with control fetuses (n = 12). At 132 ± 1 days of gestation, biceps femoris muscles were collected, and the mitochondria were isolated. Mitochondria from IUGR muscle have 47% lower State 3 (Complex I-dependent) OCRs than controls, whereas State 4 (proton leak) OCRs were not different between groups. Furthermore, Complex I, but not Complex II or IV, enzymatic activity was lower in IUGR fetuses compared with controls. Proteomic analysis (n = 6/group) identified 160 differentially expressed proteins between groups, with 107 upregulated and 53 downregulated mitochondria proteins in IUGR fetuses compared with controls. Although no differences were identified in ETC subunit protein abundances, abundances of key TCA cycle enzymes [isocitrate dehydrogenase (NAD+) 3 noncatalytic subunit ß (IDH3B), succinate-CoA ligase ADP-forming subunit-ß (SUCLA2), and oxoglutarate dehydrogenase (OGDH)] were lower in IUGR mitochondria. IUGR mitochondria had a greater abundance of a hypoxia-inducible protein, NADH dehydrogenase 1α subcomplex 4-like 2, which is known to incorporate into Complex I and lower Complex I-mediated NADH oxidation. Our findings show that mitochondria from IUGR skeletal muscle adapt to hypoxemia and hypoglycemia by lowering Complex I activity and TCA cycle enzyme concentrations, which together, act to lower OCR and NADH production/oxidation in IUGR skeletal muscle.


Asunto(s)
Ciclo del Ácido Cítrico/fisiología , Complejo I de Transporte de Electrón/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Mitocondrias Musculares/metabolismo , Animales , Regulación hacia Abajo , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Retardo del Crecimiento Fetal/enzimología , Músculos Isquiosurales/enzimología , Músculos Isquiosurales/metabolismo , Hipoglucemia/enzimología , Hipoglucemia/metabolismo , Hipoxia/enzimología , Hipoxia/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Mitocondrias Musculares/enzimología , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Consumo de Oxígeno , Insuficiencia Placentaria/enzimología , Insuficiencia Placentaria/metabolismo , Embarazo , Proteómica , Ovinos , Succinato-CoA Ligasas/metabolismo , Regulación hacia Arriba
5.
Biol Reprod ; 103(1): 126-134, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32342097

RESUMEN

The enzyme heme oxygenase-1 (HO-1), encoded by the HMOX1 gene, mediates heme catabolism by cleaving free heme. We have previously revealed the importance of HO-1 in pregnancy. Here, we determined the impact of maternal or paternal HO-1 deficiency on fetal growth and placental parameters throughout gestation. We mated Hmox1-sufficient (WT), partial (HET)-, or total (KO)-deficient BALB/c female mice with Hmox1-WT or -KO BALB/c males and performed ultrasound analysis to monitor placental and fetal growth. Doppler measurements were used to determine maternal blood flow parameters. Offspring weights and feto-placental indices (FPI) were also determined. We found a significantly increased number of underdeveloped fetuses at gd10 in HET females that were mated with WT males compared with WT × WT pairings. At the same gestational age, underdeveloped placentas could be detected in HET females mated with KO males. Many fetuses from the KO × KO combination died in utero between gd12 and gd14. At gd14, abnormal placental parameters were found in surviving fetuses, which had significant reduced weights. Moreover, only 3.11% female and 5.33% male KO pups resulted from 10 HET × HET breeding pairs over 1 year. Our results show that HO-1 from both maternal and paternal origins is important for proper placental and fetal growth. Placental growth restriction and occurrence of abortions in mice that were partially or totally deficient in HO-1 were recorded in vivo from gd10 onwards. Future studies will focus on elucidating the cellular and molecular mechanisms behind these observations.


Asunto(s)
Anemia Hemolítica/complicaciones , Retardo del Crecimiento Fetal/diagnóstico por imagen , Retardo del Crecimiento Fetal/enzimología , Edad Gestacional , Trastornos del Crecimiento/complicaciones , Hemo-Oxigenasa 1/deficiencia , Trastornos del Metabolismo del Hierro/complicaciones , Ultrasonografía Prenatal , Anemia Hemolítica/genética , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Muerte Fetal , Desarrollo Fetal/genética , Trastornos del Crecimiento/genética , Hemo-Oxigenasa 1/genética , Trastornos del Metabolismo del Hierro/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Placenta/fisiopatología , Embarazo
6.
Medicina (Kaunas) ; 55(5)2019 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-31137561

RESUMEN

Background and objectives: Intrauterine growth restriction is thought to be implicated in long-term programming of hypothalamic-pituitary-adrenal axis activity. We investigated adrenal function in adolescents born small for gestational age (SGA) in relation to their postnatal growth and cardiovascular parameters. Materials and Methods: Anthropometric parameters, blood pressure, heart rate, dehydroepiandrosterone sulfate (DHEAS), and cortisol levels were assessed in 102 adolescents aged 11-14 years followed from birth (47 SGA and 55 born appropriate for gestational age (AGA)). Results: Mean DHEAS levels were higher in SGA adolescents with catch-up growth (SGACU+), compared with AGA. Second-year height velocity and body mass index (BMI) gain during preschool years were positively related to DHEAS levels. Morning cortisol levels and systolic and diastolic blood pressure were higher in SGA adolescents without catch-up growth (SGACU-) compared with AGA. Second-year BMI gain was inversely, and 2-12 years increase in subscapular skinfold thickness was directly associated with cortisol levels. Size at birth and postnatal growth explained 47.8% and 38.2% of variation in DHEAS and cortisol levels, respectively. Conclusion: Adrenal function in adolescence is affected by prenatal and postnatal growth: small size at birth with postnatal catch-up growth is related to higher DHEAS secretion, whereas increased cortisol levels and blood pressure are higher in short SGA adolescents.


Asunto(s)
Glándulas Suprarrenales/enzimología , Retardo del Crecimiento Fetal/enzimología , Crecimiento y Desarrollo/fisiología , Adolescente , Glándulas Suprarrenales/metabolismo , Índice de Masa Corporal , Niño , Preescolar , Estudios de Cohortes , Deshidroepiandrosterona/análisis , Deshidroepiandrosterona/sangre , Femenino , Humanos , Hidrocortisona/análisis , Hidrocortisona/sangre , Lactante , Recién Nacido Pequeño para la Edad Gestacional/crecimiento & desarrollo , Modelos Lineales , Masculino , Factores Sexuales
7.
Placenta ; 81: 9-17, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31138432

RESUMEN

OBJECTIVE: Intrauterine growth restriction (IUGR) is a complication of pregnancy that has both short- and long-term sequelae for affected mothers and offspring. The pathophysiology of disease stems from poor nutrient and oxygen provision to the fetus, resulting in increased oxidative stress within the placenta. As the milieu within the local microenvironment alters macrophage differentiation, we hypothesized that macrophage plasticity may be altered in placentas associated with IUGR, and that macrophages would show hallmarks of lipid peroxidation including altered aldehyde metabolism. METHODS: In human placentas taken from normal pregnancies resulting in appropriate-for-gestational-age (AGA) newborns and placentas associated with IUGR, placental macrophages were evaluated by immunohistochemistry and shown in IUGR to resemble pro-inflammatory activated M1-type macrophages. To link oxidative stress to macrophages, the expression of aldehyde dehydrogenase (ALDHs) isozymes ALDH1, ALDH2, and ALDH3 was assessed. RESULTS: All three isozymes displayed preferential staining for distinct cellular populations within the term human placenta. ALDH1 and ALDH2 were strongly expressed in placental Hofbauer and decidual stromal cells. ALDH3, in contrast, was present in extravillous trophoblasts. Comparing AGA and IUGR-associated placentas, ALDH1 and ALDH2 trended to have greater expression in macrophage populations but lower expression in decidual cell populations in IUGR-associated placentas. ALDH3 had higher expression in IUGR-associated placentas but localized specifically to extravillous trophoblast populations. CONCLUSION: Therefore, we speculate that specific ALDH isozymes have cell-specific functions related to differentiation, inflammation, or oxidative stress responses that are altered in IUGR-associated term human placentas. This family of isozymes may be a novel method to identify human placentas affected by placental insufficiency/IUGR.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Retardo del Crecimiento Fetal/enzimología , Macrófagos/metabolismo , Placenta/enzimología , Adulto , Femenino , Retardo del Crecimiento Fetal/inmunología , Humanos , Embarazo , Isoformas de Proteínas/metabolismo
8.
Prenat Diagn ; 39(1): 3-9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30474229

RESUMEN

Published studies indicate the MTHFR C677T and A1298C polymorphisms are associated with abnormal homocysteine levels, which may cause various pregnancy complications and birth defects. However, the results obtained from different studies have been inconsistent. Therefore, this meta-analysis explores the association between MTHFR polymorphisms and birth defects and adverse pregnancy outcomes. The PubMed, ScienceDirect, Embase, and China Biology Medicine literature databases and ClinicalTrials were searched. Analyses of public bias, meta-regression, subgroups, and sensitivity were used to ensure the robustness of our results. MTHFR C677T was significantly associated with recurrent pregnancy loss in developing countries (odds ratio [OR], 1.34; 95% confidence interval [CI], 1.20-1.50) but not in developed countries (OR, 0.87; 95% CI, 0.68-1.11). No significant relationship was found between MTHFR A1298C and recurrent pregnancy loss (OR, 1.04; 95% CI, 0.93-1.18). MTHFR C677T and A1298C were not associated with preeclampsia (OR, 1.06; 95% CI, 0.97-1.16 and OR, 1.16; 95% CI, 0.97-1.39, respectively), and C677T was not associated with placental abruption (OR, 1.03; 95% CI, 0.87-1.21), intrauterine growth retardation (OR, 1.02; 95% CI, 0.90-1.15), or congenital heart disease (OR, 1.05; 95% CI, 0.89-1.25). MTHFR C677T, but not A1298C, was associated with neural tube defects (OR, 1.24; 95% CI, 1.08-1.42) and Down syndrome (OR, 1.65; 95% CI, 1.39-1.95). CONCLUSION: Although MTHFR C677T and A1298C are significantly associated with some types of congenital defects and adverse pregnancy outcomes, the impact of these polymorphisms is moderate.


Asunto(s)
Anomalías Congénitas/enzimología , Anomalías Congénitas/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Resultado del Embarazo/genética , Aborto Habitual/enzimología , Aborto Habitual/genética , Ensayos Clínicos como Asunto , Países en Desarrollo , Síndrome de Down/enzimología , Síndrome de Down/genética , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/genética , Humanos , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/genética , Oportunidad Relativa , Embarazo
9.
Endocrinology ; 160(2): 377-386, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535296

RESUMEN

Obesity and unhealthy nutrition are increasing and affect women of childbearing age and hence during pregnancy. Despite normal or even high birth weight, the offspring suffers from long-term metabolic risks. We hypothesized that fetal growth is disturbed during different intrauterine phases. Underlying molecular events remain elusive. Female mice were fed either a standard diet (SD) or a high-fat diet (HFD) after weaning until mating and during pregnancy. Pregnant mice were euthanized at gestational day (G)15.5 and G18.5, and fetuses and placentas were removed for analysis. HFD fetuses displayed intrauterine growth restriction (IUGR) at G15.5, which disappeared until G18.5, indicating intrauterine catch-up growth during that time period. Main placental findings indicate decreased canonical Wnt-GSK3ß signaling and lower proliferation rates at G18.5, which goes along with a smaller placental transfer zone. On the other hand, glucose depots (glycogen cluster) in HFD placentas decreased more strongly between G15.5 and G18.5 compared with placentas from SD mothers, and the glucose transporter protein GLUT-1 was increased at G18.5 in the HFD group. Maternal diet-induced obesity causes an IUGR phenotype at the beginning of the third week (G15.5) in our mouse model. This phenotype is reversed by the end of the third week (G18.5) despite a smaller placental transfer zone, probably based on GSK3ß-mediated increased glucose mobilization in the placenta and hence an increased glucose supply to the fetus.


Asunto(s)
Desarrollo Fetal , Retardo del Crecimiento Fetal/etiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Obesidad/fisiopatología , Placenta/metabolismo , Animales , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/fisiopatología , Masculino , Ratones , Obesidad/enzimología , Placenta/fisiopatología , Embarazo
10.
PLoS One ; 13(9): e0203802, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30212527

RESUMEN

Antenatal overexposure to glucocorticoids causes fetal intrauterine growth restriction (IUGR) and adult metabolic disorders. 11ß-hydroxysteroid dehydrogenase (11ß-HSD) 1 and 2 are key enzymes for glucocorticoid metabolism, however, the detailed effects of antenatal overexposure to glucocorticoids on placental 11ß-HSD1 and 2 expression and adult metabolic disorders remain obscure. Here, we report that, in placenta 11ß-HSD1 is diffusely localized, whereas 11ß-HSD2 is specifically expressed in labyrinthine layer. Exposure of pregnant dams to betamethasone significantly increases the expression of placental 11ß-HSD2 but not 11ß-HSD1, and decreases the weights of fetuses but not placentas. Antenatal exposure to betamethasone leads to either significant weight loss in the offspring younger than 10-week-old, or weight gain in those older than 14-week-old. Furthermore, antenatal exposure to betamethasone results in coexistence of various metabolic disorders in adult offspring, including hyperglycemia, glucose intolerance, low insulin secretory capacity and hyperlipidemia. The present study demonstrates that exposure of pregnant dams to betamethasone induces the expression of placental 11ß-HSD2 but not 11ß-HSD1, leads to fetal IUGR and causes adult metabolic disorders, providing evidence for fetal origins of adult diseases and the potential role of placental 11ß-HSD2 in them.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Betametasona/efectos adversos , Glucocorticoides/efectos adversos , Placenta/efectos de los fármacos , Placenta/enzimología , Efectos Tardíos de la Exposición Prenatal , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/enzimología , Glucosa/metabolismo , Insulina/metabolismo , Lípidos/sangre , Masculino , Enfermedades Metabólicas/inducido químicamente , Enfermedades Metabólicas/enzimología , Ratones Endogámicos ICR , Placenta/fisiología , Embarazo
11.
Pharmacol Res ; 134: 68-78, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29890254

RESUMEN

The effects of intrauterine growth restriction (IUGR) extend well into postnatal life. IUGR is associated with an increased risk of adverse health outcomes, which often leads to greater medication usage. Many medications require hepatic metabolism for activation or clearance, but hepatic function may be altered in IUGR fetuses. Using a sheep model of IUGR, we determined the impact of IUGR on hepatic drug metabolism and drug transporter expression, both important mediators of fetal drug exposure, in late gestation and in neonatal life. In the late gestation fetus, IUGR decreased the gene expression of uptake drug transporter OATPC and increased P-glycoprotein protein expression in the liver, but there was no change in the activity of the drug metabolising enzymes CYP3A4 or CYP2D6. In contrast, at 3 weeks of age, CYP3A4 activity was reduced in the livers of lambs born with low birth weight (LBW), indicating that LBW results in changes to drug metabolising capacity in neonatal life. Together, these results suggest that IUGR may reduce hepatic drug metabolism in fetal and neonatal life through different mechanisms.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Retardo del Crecimiento Fetal/enzimología , Hígado/enzimología , Transportadores de Anión Orgánico/metabolismo , Preparaciones Farmacéuticas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Animales Recién Nacidos , Peso al Nacer , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP3A/genética , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/genética , Peso Fetal , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Transportadores de Anión Orgánico/genética , Embarazo , Oveja Doméstica
12.
Am J Physiol Regul Integr Comp Physiol ; 315(3): R509-R520, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29741931

RESUMEN

Individuals born after intrauterine growth restriction (IUGR) are at increased risk of developing cardiovascular diseases in adulthood, notably hypertension (HTN). Alterations in the vascular system, particularly impaired endothelium-dependent vasodilation, may play an important role in long-term effects of IUGR. Whether such vascular dysfunction precedes HTN has not been fully established in individuals born after IUGR. Moreover, the intimate mechanisms of altered endothelium-dependent vasodilation remain incompletely elucidated. We therefore investigated, using a rat model of IUGR, whether impaired endothelium-dependent relaxation precedes the development of HTN and whether key components of the l-arginine-nitric oxide (NO) pathway are involved in its pathogenesis. Pregnant rats were fed with a control (CTRL, 23% casein) or low-protein diet (LPD, 9% casein) to induce IUGR. Systolic blood pressure (SBP) was measured by tail-cuff plethysmography in 5- and 8-wk-old male offspring. Aortic rings were isolated to investigate relaxation to acetylcholine, NO production, endothelial NO synthase (eNOS) protein content, arginase activity, and superoxide anion production. SBP was not different at 5 wk but significantly increased in 8-wk-old offspring of maternal LPD (LP) versus CTRL offspring. In 5-wk-old LP versus CTRL males, endothelium-dependent vasorelaxation was significantly impaired but restored by preincubation with l-arginine or the arginase inhibitor S-(2-boronoethyl)-l-cysteine; NO production was significantly reduced but restored by l-arginine pretreatment; total eNOS protein, dimer-to-monomer ratio, and arginase activity were significantly increased; superoxide anion production was significantly enhanced but normalized by pretreatment with the NO synthase inhibitor Nω-nitro-l-arginine. In this model, IUGR leads to early-impaired endothelium-dependent vasorelaxation, resulting from arginase upregulation and eNOS uncoupling, which precedes the development of HTN.


Asunto(s)
Aorta Torácica/enzimología , Arginasa/metabolismo , Endotelio Vascular/enzimología , Retardo del Crecimiento Fetal/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Vasodilatación , Factores de Edad , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Aorta Torácica/fisiopatología , Arginina/metabolismo , Dieta con Restricción de Proteínas , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Femenino , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/fisiopatología , Hipertensión/enzimología , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Óxido Nítrico/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas Sprague-Dawley , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
13.
Sci Rep ; 8(1): 5488, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29615752

RESUMEN

Indoleamine 2,3-dioxygenase-1 (IDO1) mediates the degradation of L-tryptophan (L-Trp) and is constitutively expressed in the chorionic vascular endothelium of the human placenta with highest levels in the microvasculature. Given that endothelial expression of IDO1 has been shown to regulate vascular tone and blood pressure in mice under the condition of systemic inflammation, we asked whether IDO1 is also involved in the regulation of placental blood flow and if yes, whether this function is potentially impaired in intrauterine growth restriction (IUGR) and pre-eclampsia (PE). In the large arteries of the chorionic plate L-Trp induced relaxation only after upregulation of IDO1 using interferon gamma and tumor necrosis factor alpha. However, ex vivo placental perfusion of pre-constricted cotyledonic vasculature with L-Trp decreases the vessel back pressure without prior IDO1 induction. Further to this finding, IDO1 protein expression and activity is reduced in IUGR and PE when compared to gestational age-matched control tissue. These data suggest that L-Trp catabolism plays a role in the regulation of placental vascular tone, a finding which is potentially linked to placental and fetal growth. In this context our data suggest that IDO1 deficiency is related to the pathogenesis of IUGR and PE.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Endotelio Vascular/enzimología , Retardo del Crecimiento Fetal/enzimología , Placenta/irrigación sanguínea , Preeclampsia/enzimología , Adulto , Arterias/fisiopatología , Endotelio Vascular/metabolismo , Femenino , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Regulación Enzimológica de la Expresión Génica , Humanos , Preeclampsia/patología , Preeclampsia/fisiopatología , Embarazo , Vasodilatación
14.
Pregnancy Hypertens ; 11: 7-11, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29523277

RESUMEN

OBJECTIVE: The enzyme 11ß-dehydroxysteroid dehydrogenase 2 (11ß-HSD2) converts active cortisol (F) to inactive cortisone (E). A reduced 11ß-HSD2 activity in the placenta has been demonstrated for prematurity, low birth weight, and preeclampsia. We hypothesized that disturbed placental function rather than a maternal response contributes to decreased 11ßHSD2 activity as reflected by a diminished conversion of F to E. Hence, the aim of the present study was to estimate the systemic activity of 11ß-HSD2 throughout gestation and in pregnancies complicated by preeclampsia (PE) and intrauterine growth restriction (IUGR) by calculating maternal serum F/E ratios. METHODS: A total of 188 maternal serum samples were analyzed for nine glucocorticoid metabolites by gas chromatography-mass spectrometry (GC-MS) and F/E ratios were calculated. Study Group A: In a longitudinal set 33 healthy pregnant women were analyzed at three different time points throughout gestation and one postpartum. Study Group B: Cross-sectionally additional 56 patients were enrolled. We compared patients with PE (N = 14) and IUGR (N = 14) with gestational age matched healthy controls (CTRL = 28). RESULTS: Group A: The apparent 11ß-HSD2 activity dropped in the second trimester being restored to first trimester levels (P value = 0.016). Group B: The 11ß-HSD2 activity was high in PE (P value < 0.05) but not in the IUGR group as compared to CTRL. CONCLUSION: The increased apparent serum 11ß-HSD2 activity observed with advancing gestation in normal pregnancy may reflect an elevated general increase in enzyme activity due to a higher placental mass. The high systemic 11ß-HSD2 activity in PE but not in IUGR however suggests an increased F deactivation in maternal tissue in PE rather than in the placenta since placental insufficiency in the absence of PE does not significantly alter F/E ratio.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/sangre , Retardo del Crecimiento Fetal/sangre , Preeclampsia/sangre , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Cortisona/sangre , Estudios Transversales , Femenino , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/enzimología , Edad Gestacional , Humanos , Hidrocortisona/sangre , Estudios Longitudinales , Preeclampsia/diagnóstico , Preeclampsia/enzimología , Embarazo , Regulación hacia Arriba
15.
J Matern Fetal Neonatal Med ; 30(10): 1200-1206, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27380314

RESUMEN

OBJECTIVE: Recent evidence suggests that oxidative stress is involved in the pathophysiology of many human diseases. It has been demonstrated that oxidative stress is associated with intrauterine growth restriction (IUGR), and the depletion of placental antioxidant systems has been suggested as a key factor in this disease. Our aims were to explore the possible role of antioxidant paraoxonase-2 (PON2) and paraoxonase-3 (PON3) in the pathophysiology of unexplained IUGR. METHODS: We have studied the expression of mRNA for PON2, PON3 in placental tissues by using RT-qPCR. Two groups, consisting of normal (n = 18) and unexplained IUGR pregnancies (n = 20) were compared. RESULTS: Our results demonstrated that there were no significant differences in the mRNA expressions of PON2, PON3 between the two groups (p = 0.28, p = 0.90, respectively). PON2 and PON3 were down-regulated in IUGR. Antenatal steroid therapy had no effect on the expression mRNA in placentae of unexplained IUGR pregnancies compared to non-treated group. CONCLUSIONS: These results suggest that PON2, PON3 mRNA levels were not changed significantly in placentae of IUGR when compared to normal pregnant women.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Retardo del Crecimiento Fetal/enzimología , Placenta/enzimología , Adulto , Arildialquilfosfatasa/genética , Estudios de Casos y Controles , Femenino , Retardo del Crecimiento Fetal/etiología , Humanos , Estrés Oxidativo , Embarazo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Exp Suppl ; 107: 73-79, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27812977

RESUMEN

AMPK is important in numerous physiological systems but plays a vital role in embryonic and placental development. The placenta is a unique organ that is the essential lifeline between the mother and baby during pregnancy and gestation. During placental development, oxygen concentrations are very low until cells differentiate to establish the appropriate lineages that take on new functions required for placental and embryonic survival. Balancing the oxygen regulatory environment with the demands for energy and need to maintain metabolism during this process places AMPK at the center of maintaining placental cellular homeostasis as it integrates and responds to numerous complex stimuli. AMPK plays a critical role in sensing metabolic and energy changes. Once activated, it turns on pathways that produce energy and shuts down catabolic processes. AMPK coordinates cell growth, differentiation, and nutrient transport to maintain cell survival. Appropriate regulation of AMPK is essential for normal placental and embryonic development, and its dysregulation may lead to pregnancy-associated disorders such as intrauterine growth restriction, placental insufficiency, or preeclampsia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Oxígeno/metabolismo , Placenta/enzimología , Placentación/genética , Células Madre/enzimología , Proteínas Quinasas Activadas por AMP/metabolismo , Diferenciación Celular , Proliferación Celular , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/patología , Regulación de la Expresión Génica , Humanos , Placenta/citología , Insuficiencia Placentaria/enzimología , Insuficiencia Placentaria/etiología , Insuficiencia Placentaria/genética , Insuficiencia Placentaria/patología , Preeclampsia/enzimología , Preeclampsia/etiología , Preeclampsia/genética , Preeclampsia/patología , Embarazo , Complicaciones del Embarazo/enzimología , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/patología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transducción de Señal , Células Madre/patología
17.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1226-43, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26936783

RESUMEN

Intrauterine growth restriction induced by placental restriction (PR) in sheep leads to chronic hypoxemia and reduced surfactant maturation. The underlying molecular mechanism involves altered regulation of hypoxia signaling by increased prolyl hydroxylase domain (PHD) expression. Here, we evaluated the effect of intratracheal administration of the PHD inhibitor dimethyloxalylglycine (DMOG) on functional, molecular, and structural determinants of lung maturation in the control and PR sheep fetus. There was no effect of DMOG on fetal blood pressure or fetal breathing movements. DMOG reduced lung expression of genes regulating hypoxia signaling (HIF-3α, ACE1), antioxidant defense (CAT), lung liquid reabsorption (SCNN1-A, ATP1-A1, AQP-1, AQP-5), and surfactant maturation (SFTP-A, SFTP-B, SFTP-C, PCYT1A, LPCAT, ABCA3, LAMP3) in control fetuses. There were very few effects of DMOG on gene expression in the PR fetal lung (reduced lung expression of angiogenic factor ADM, water channel AQP-5, and increased expression of glucose transporter SLC2A1). DMOG administration in controls reduced total lung lavage phosphatidylcholine to the same degree as in PR fetuses. These changes appear to be regulated at the molecular level as there was no effect of DMOG on the percent tissue, air space, or numerical density of SFTP-B positive cells in the control and PR lung. Hence, DMOG administration mimics the effects of PR in reducing surfactant maturation in the lung of control fetuses. The limited responsiveness of the PR fetal lung suggests a potential biochemical limit or reduced plasticity to respond to changes in regulation of hypoxia signaling following exposure to chronic hypoxemia in utero.


Asunto(s)
Retardo del Crecimiento Fetal/enzimología , Pulmón/enzimología , Pulmón/crecimiento & desarrollo , Prolil Hidroxilasas/metabolismo , Inhibidores de Prolil-Hidroxilasa/metabolismo , Animales , Femenino , Edad Gestacional , Pulmón/embriología , Prolil Hidroxilasas/química , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo , Ovinos , Relación Estructura-Actividad
18.
PLoS One ; 11(3): e0151617, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26974824

RESUMEN

Preeclampsia (PE) is a potentially fatal pregnancy-related hypertensive disorder characterized by poor placenta development that can cause fetal growth restriction. PE-associated pathologies, including thrombosis, hypertension, and impaired placental development, may result from imbalances between thromboxane A2 (TXA2) and prostacyclin. Low-dose aspirin, which selectively inhibits TXA2 production, is used to prevent high-risk PE. However, the role of TXA2 in aspirin-mediated protective effects in women with PE is not understood fully. In this study, we examined the role of prostanoids in PE using human samples and an induced PE mouse model. We demonstrated that the administration of salted drinking water (2.7% NaCl) to wild-type mice resulted in elevated placental TXA2 synthase (TXAS) and plasma TXA2, but not prostacyclin, levels, which was also found in our clinical PE placenta samples. The high salt-treated wild-type pregnant mice had shown unchanged maternal body weight, hypertension (MAP increase 15 mmHg), and decreased pup weight (~50%) and size (~24%), but these adverse effects were ameliorated in TXAS knockout (KO) mice. Moreover, increased expression of interleukin-1ß and downstream phosphorylated-p38-mitogen-activated protein kinase were concordant with apoptosis induction in the placentas of salt water-treated wild-type mice. These alterations were not observed in TXAS KO mice. Together, our data suggest that TXA2 depletion has anti-PE effects due to the prevention of hypertension and placental damage through downregulation of the interleukin-1ß pathway.


Asunto(s)
Retardo del Crecimiento Fetal/prevención & control , Hipertensión/prevención & control , Cloruro de Sodio Dietético/efectos adversos , Tromboxano-A Sintasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Recuento de Células , Cloruros/sangre , Femenino , Retardo del Crecimiento Fetal/enzimología , Humanos , Hipertensión/sangre , Hipertensión/complicaciones , Hipertensión/enzimología , Interleucina-1beta/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Perfusión , Fenotipo , Placenta/metabolismo , Preeclampsia/sangre , Preeclampsia/enzimología , Embarazo , Transducción de Señal/efectos de los fármacos , Sodio/sangre , Regulación hacia Arriba
19.
Toxicology ; 340: 43-52, 2016 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-26776438

RESUMEN

Prenatal nicotine exposure is a risk factor for intrauterine growth retardation (IUGR). Steroid hormones synthesized from cholesterol in the fetal adrenal play an important role in the fetal development. The aim of this study is to investigate the effects of prenatal nicotine exposure on steroidogenesis in fetal rat adrenals from the perspective of cholesterol supply and explore the underlying epigenetic mechanisms. Pregnant Wistar rats were administered 1.0mg/kg nicotine subcutaneously twice a day from gestational day (GD) 7 to GD17. The results showed that prenatal nicotine exposure increased IUGR rates. Histological changes, decreased steroid hormone concentrations and decreased cholesterol supply were observed in nicotine-treated fetal adrenals. In the gene expression array, the expression of genes regulating ketone metabolic process decreased in nicotine-treated fetal adrenals. The following conjoint analysis of DNA methylation array with these differentially expressed genes suggested that acetoacetyl-CoA synthetase (AACS), the enzyme utilizing ketones for cholesterol supply, may play an important role in nicotine-induced cholesterol supply deficiency. Moreover, the decreased expression of AACS and increased DNA methylation in the proximal promoter of AACS in the fetal adrenal was verified by real-time reverse-transcription PCR (RT-PCR) and bisulfite sequencing PCR (BSP), respectively. In conclusion, prenatal nicotine exposure can cause DNA hypermethylation of the AACS promoter in the rat fetal adrenal. These changes may result in decreased AACS expression and cholesterol supply, which inhibits steroidogenesis in the fetal adrenal.


Asunto(s)
Corticoesteroides/biosíntesis , Glándulas Suprarrenales/efectos de los fármacos , Colesterol/metabolismo , Coenzima A Ligasas/metabolismo , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Retardo del Crecimiento Fetal/inducido químicamente , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Glándulas Suprarrenales/embriología , Glándulas Suprarrenales/enzimología , Animales , Coenzima A Ligasas/genética , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/genética , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Exposición Materna , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas Wistar
20.
Cell Rep ; 13(12): 2817-28, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26711346

RESUMEN

Maternal immune cells are an integral part of reproduction, but how they might cause pregnancy complications remains elusive. Macrophages and their dual function in inflammation and tissue repair are thought to play key yet undefined roles. Altered perinatal growth underpins adult morbidity, and natural killer (NK) cells may sustain fetal growth by establishing the placental blood supply. Using a mouse model of genetic inactivation of PI3K p110δ, a key intracellular signaling molecule in leukocytes, we show that p110δ regulates macrophage dynamics and NK-cell-mediated arterial remodeling. The uterus of dams with inactive p110δ had decreased IFN-γ and MHC class II(low) macrophages but enhanced IL-6. Poor vascular remodeling and a pro-inflammatory uterine milieu resulted in fetal death or growth retardation. Our results provide one mechanism that explains how imbalanced adaptations of maternal innate immune cells to gestation affect offspring well-being with consequence perinatally and possibly into adulthood.


Asunto(s)
Muerte Fetal , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Animales , Fosfatidilinositol 3-Quinasa Clase I , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/inmunología , Silenciador del Gen , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Macrófagos/inmunología , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Transducción de Señal , Útero/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...