Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 226
Filtrar
1.
Clin Sci (Lond) ; 138(13): 761-775, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38904187

RESUMEN

Placental insufficiency is one of the major causes of fetal growth restriction (FGR), a significant pregnancy disorder in which the fetus fails to achieve its full growth potential in utero. As well as the acute consequences of being born too small, affected offspring are at increased risk of cardiovascular disease, diabetes and other chronic diseases in later life. The placenta and heart develop concurrently, therefore placental maldevelopment and function in FGR may have profound effect on the growth and differentiation of many organ systems, including the heart. Hence, understanding the key molecular players that are synergistically linked in the development of the placenta and heart is critical. This review highlights the key growth factors, angiogenic molecules and transcription factors that are common causes of defective placental and cardiovascular development.


Asunto(s)
Retardo del Crecimiento Fetal , Placenta , Humanos , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/fisiopatología , Embarazo , Femenino , Placenta/metabolismo , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/fisiopatología , Animales , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/embriología , Sistema Cardiovascular/fisiopatología , Sistema Cardiovascular/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intercelular/metabolismo
2.
Nature ; 629(8012): 652-659, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693261

RESUMEN

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Asunto(s)
Susceptibilidad a Enfermedades , Disbiosis , Padre , Microbioma Gastrointestinal , Insuficiencia Placentaria , Lesiones Prenatales , Espermatozoides , Animales , Femenino , Masculino , Ratones , Embarazo , Disbiosis/complicaciones , Disbiosis/microbiología , Microbioma Gastrointestinal/fisiología , Leptina/metabolismo , Ratones Endogámicos C57BL , Placenta/metabolismo , Placenta/fisiopatología , Insuficiencia Placentaria/etiología , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/fisiopatología , Resultado del Embarazo , Lesiones Prenatales/etiología , Lesiones Prenatales/metabolismo , Lesiones Prenatales/fisiopatología , Transducción de Señal , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/fisiopatología , Susceptibilidad a Enfermedades/etiología
3.
Placenta ; 151: 19-25, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38657321

RESUMEN

INTRODUCTION: Placental insufficiency may lead to preeclampsia and fetal growth restriction. There is no cure for placental insufficiency, emphasizing the need for monitoring fetal and placenta health. Current monitoring methods are limited, underscoring the necessity for imaging techniques to evaluate fetal-placental perfusion and oxygenation. This study aims to use MRI to evaluate placental oxygenation and perfusion in the reduced uterine perfusion pressure (RUPP) model of placental insufficiency. METHODS: Pregnant rats were randomized to RUPP (n = 11) or sham surgery (n = 8) on gestational day 14. On gestational day 19, rats imaged using a 7T MRI scanner to assess oxygenation and perfusion using T2* mapping and 3D-DCE MRI sequences, respectively. The effect of the RUPP on the feto-placental units were analyzed from the MRI images. RESULTS: RUPP surgery led to reduced oxygenation in the labyrinth (24.7 ± 1.8 ms vs. 28.0 ± 2.1 ms, P = 0.002) and junctional zone (7.0 ± 0.9 ms vs. 8.1 ± 1.1 ms, P = 0.04) of the placenta, as indicated by decreased T2* values. However, here were no significant differences in fetal organ oxygenation or placental perfusion between RUPP and sham animals. DISCUSSION: The reduced placental oxygenation without a corresponding decrease in perfusion suggests an adaptive response to placental ischemia. While acute reduction in placental perfusion may cause placental hypoxia, persistence of this condition could indicate chronic placental insufficiency after ischemic reperfusion injury. Thus, placental oxygenation may be a more reliable biomarker for assessing fetal condition than perfusion in hypertensive disorders of pregnancies including preeclampsia and FGR.


Asunto(s)
Modelos Animales de Enfermedad , Imagen por Resonancia Magnética , Oxígeno , Placenta , Insuficiencia Placentaria , Ratas Sprague-Dawley , Animales , Embarazo , Femenino , Insuficiencia Placentaria/diagnóstico por imagen , Insuficiencia Placentaria/metabolismo , Imagen por Resonancia Magnética/métodos , Placenta/diagnóstico por imagen , Placenta/metabolismo , Placenta/irrigación sanguínea , Ratas , Oxígeno/metabolismo , Circulación Placentaria/fisiología , Imagenología Tridimensional/métodos , Medios de Contraste
4.
Int J Mol Sci ; 25(3)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38338665

RESUMEN

We report the case of a four-year-old male patient with a complex medical history born prematurely as the result of intrauterine growth restriction due to placental insufficiency. His clinical manifestations included severe neurodevelopmental deficits, global developmental delay, Pierre-Robin sequence, and intractable epilepsy with both generalized and focal features. The proband's low levels of citrulline and lactic acidosis provoked by administration of Depakoke were evocative of a mitochondrial etiology. The proband's genotype-phenotype correlation remained undefined in the absence of nuclear and mitochondrial pathogenic variants detected by deep sequencing of both genomes. However, live-cell mitochondrial metabolic investigations provided evidence of a deficient oxidative-phosphorylation pathway responsible for adenosine triphosphate (ATP) synthesis, leading to chronic energy crisis in the proband. In addition, our metabolic analysis revealed metabolic plasticity in favor of glycolysis for ATP synthesis. Our mitochondrial morphometric analysis by transmission electron microscopy confirmed the suspected mitochondrial etiology, as the proband's mitochondria exhibited an immature morphology with poorly developed and rare cristae. Thus, our results support the concept that suboptimal levels of intrauterine oxygen and nutrients alter fetal mitochondrial metabolic reprogramming toward oxidative phosphorylation (OXPHOS) leading to a deficient postnatal mitochondrial energy metabolism. In conclusion, our collective studies shed light on the long-term postnatal mitochondrial pathophysiology caused by intrauterine growth restriction due to idiopathic placental insufficiency and its negative impact on the energy-demanding development of the fetal and postnatal brain.


Asunto(s)
Retardo del Crecimiento Fetal , Insuficiencia Placentaria , Masculino , Humanos , Femenino , Embarazo , Preescolar , Retardo del Crecimiento Fetal/metabolismo , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/patología , Placenta/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo
5.
Pediatr Res ; 95(6): 1410-1411, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38263448

RESUMEN

Uteroplacental insufficiency (UPI) is a major cause of fetal growth restriction (FGR). Leptin, an adipokine, has been shown to play a vital role in fetal organogenesis. There is evidence reporting leptin deficiency in preterm and growth-restricted fetuses. In this issue of Pediatric Research, Yuliana et al. report leptin expression and lung development in UPI-induced FGR rats. UPI-induced FGR rats expressed decreased lung leptin and had impaired lung development, as shown by decreased surface area and lung volume. They also found a significant association between lung radial alveolar count, serum leptin, von Willebrand factor, and specific metabolites on metabolomic analyses. Previous studies on leptin supplementation in vivo have been associated with improvement in lung maturation; supporting the evidence, that leptin improves lung growth and development in FGR and may have future therapeutic potential in the improvement of respiratory outcomes in these infants. Future studies to support evidence of this association in humans are warranted.


Asunto(s)
Retardo del Crecimiento Fetal , Leptina , Pulmón , Insuficiencia Placentaria , Animales , Femenino , Humanos , Embarazo , Ratas , Desarrollo Fetal , Retardo del Crecimiento Fetal/metabolismo , Leptina/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Insuficiencia Placentaria/metabolismo
6.
Endocrinology ; 165(1)2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38035825

RESUMEN

Prevailing hypoxemia and hypoglycemia in near-term fetuses with placental insufficiency-induced intrauterine growth restriction (IUGR) chronically increases norepinephrine concentrations, which lower adrenergic sensitivity and lipid mobilization postnatally, indicating a predisposition for adiposity. To determine adrenergic-induced responses, we examined the perirenal adipose tissue transcriptome from IUGR fetuses with or without hypercatecholaminemia. IUGR was induced in sheep with maternal hyperthermia, and hypercatecholaminemia in IUGR was prevented with bilateral adrenal demedullation. Adipose tissue was collected from sham-operated control (CON) and IUGR fetuses and adrenal-demedullated control (CAD) and IUGR (IAD) fetuses. Norepinephrine concentrations were lower in IAD fetuses than in IUGR fetuses despite both being hypoxemic and hypoglycemic. In IUGR fetuses, perirenal adipose tissue mass relative to body mass was greater compared with the CON, adrenal-demedullated control, and IAD groups. Transcriptomic analysis identified 581 differentially expressed genes (DEGs) in CON vs IUGR adipose tissue and 193 DEGs in IUGR vs IAD adipose tissue. Integrated functional analysis of these 2 comparisons showed enrichment for proliferator-activated receptor signaling and metabolic pathways and identified adrenergic responsive genes. Within the adrenergic-regulated DEGs, we identified transcripts that regulate adipocyte proliferation and differentiation: adipogenesis regulatory factor, C/CCAAT/enhancer binding protein α, and sterol carrier protein 2. DEGs associated with the metabolic pathway included pyruvate dehydrogenase kinase 4, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4, IGF-binding proteins (IGFBP-5 and IGFBP-7). Sex-specific expression differences were also found for adipogenesis regulatory factor, pyruvate dehydrogenase kinase 4, IGFBP5, and IGFBP7. These findings indicate that sustained adrenergic stimulation during IUGR leads to adipocyte hyperplasia with alterations in metabolism, proliferation, and preadipocyte differentiation pathways.


Asunto(s)
Retardo del Crecimiento Fetal , Insuficiencia Placentaria , Masculino , Humanos , Ovinos , Animales , Femenino , Embarazo , Retardo del Crecimiento Fetal/metabolismo , Norepinefrina/metabolismo , Insuficiencia Placentaria/metabolismo , Hiperplasia/metabolismo , Placenta/metabolismo , Adipocitos/metabolismo , Adrenérgicos/metabolismo , Feto/metabolismo
7.
Biol Reprod ; 109(6): 982-993, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37724932

RESUMEN

During implantation, trophoblast cell invasion and differentiation is predominantly important to achieving proper placental formation and embryonic development. The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) working through its receptor C-X-C motif chemokine receptor 4 (CXCR4) is implicated in implantation and placentation but precise roles of this axis are unclear. Suppressing CXCL12/CXCR4 signaling at the fetal-maternal interface in sheep reduces trophoblast invasion, disrupts uterine remodeling, and diminishes placental vascularization. We hypothesize these negative impacts during implantation will manifest as compromised fetal and placental growth at midgestation. To test, on day 12 postbreeding, osmotic pumps were surgically installed in 30 ewes and delivered intrauterine CXCR4 inhibitor or saline for 7 or 14 days. On day 90, fetal/maternal tissues were collected, measured, weighed, and maternal (caruncle) and fetal (cotyledon) placenta components separated and analyzed. The objectives were to determine if (i) suppressing CXCL12/CXCR4 during implantation results in reduced fetal and placental growth and development and (ii) if varying the amount of time CXCL12/CXCR4 is suppressed impacts fetal/placental development. Fetal weights were similar; however greater placental weight and placentome numbers occurred when CXCL12/CXCR4 was suppressed for 14 days. In caruncles, greater abundance of fibroblast growth factor 2, vascular endothelial growth factor A, vascular endothelial growth factor A receptor 1 (FLT-1), and placental growth factor were observed after suppressing CXCL12/CXCR4. Similar results occurred in cotyledons except less vascular endothelial growth factor in 7 day group and less fibroblast growth factor in 14 day group. Our data underscore the importance of CXCL12/CXCR4 signaling during placentation and provide strong evidence that altering CXCL12-mediated signaling induces enduring placental effects manifesting later in gestation.


Asunto(s)
Placenta , Insuficiencia Placentaria , Humanos , Embarazo , Femenino , Ovinos , Animales , Placenta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Insuficiencia Placentaria/metabolismo , Factor de Crecimiento Placentario/metabolismo , Placentación , Quimiocina CXCL12/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
8.
Placenta ; 139: 68-74, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37331027

RESUMEN

INTRODUCTION: Placental insufficiency is a leading cause of intrauterine growth restriction, contributing to perinatal morbidity and mortality. The molecular regulation of placental development and what causes placental insufficiency is poorly understood. Recently, a panel of genes were found to cause significant placental dysmorphologies in mice with severely growth restricted off-spring. We aimed to assess whether these genes were also implicated in human intrauterine growth restriction. METHODS: We explored the expression of nine genes in primary cytotrophoblast cells in hypoxic (n = 6) and glucose starvation (n = 5) conditions in vitro. We also explored whether the genes were dysregulated in intrauterine growth restricted human placental samples (n = 11), with (n = 20) or without preeclampsia compared to gestationally matched controls (<34 weeks gestation) (n = 17). RESULTS: Hypoxic stress significantly upregulated the expressions of BRD2 (p = 0.0313), SMG9 (p = 0.0313) genes. In contrast, glucose starvation significantly suppressed Kif1bp (p = 0.0089) in primary cytotrophoblasts. The FRYL, NEK9, CHTOP, PSPH, ATP11A, HM13 genes did not change under hypoxia or glucose starvation conditions. The expression of these genes was not altered in placenta from patients with intrauterine growth restriction, compared to gestationally matched controls. DISCUSSION: We demonstrate that some of the genes that cause a placental phenotype in mice, respond to hypoxic and glucose mediated stress in human cytotrophoblast isolations. Despite this, they are unchanged in placenta from patients with intrauterine growth restriction. Therefore, dysregulation of these genes is less likely to contribute to preterm intrauterine growth restriction in humans.


Asunto(s)
Insuficiencia Placentaria , Preeclampsia , Humanos , Embarazo , Femenino , Animales , Ratones , Placenta/metabolismo , Trofoblastos/metabolismo , Insuficiencia Placentaria/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Ratones Noqueados , Placentación , Hipoxia/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Fenotipo , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo
9.
Am J Physiol Endocrinol Metab ; 324(6): E556-E568, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37126847

RESUMEN

Glucose, lactate, and amino acids are major fetal nutrients. During placental insufficiency-induced intrauterine growth restriction (PI-IUGR), uteroplacental weight-specific oxygen consumption rates are maintained, yet fetal glucose and amino acid supply is decreased and fetal lactate concentrations are increased. We hypothesized that uteroplacental metabolism adapts to PI-IUGR by altering nutrient allocation to maintain oxidative metabolism. Here, we measured nutrient flux rates, with a focus on nutrients shuttled between the placenta and fetus (lactate-pyruvate, glutamine-glutamate, and glycine-serine) in a sheep model of PI-IUGR. PI-IUGR fetuses weighed 40% less and had decreased oxygen, glucose, and amino acid concentrations and increased lactate and pyruvate versus control (CON) fetuses. Uteroplacental weight-specific rates of oxygen, glucose, lactate, and pyruvate uptake were similar. In PI-IUGR, fetal glucose uptake was decreased and pyruvate output was increased. In PI-IUGR placental tissue, pyruvate dehydrogenase (PDH) phosphorylation was decreased and PDH activity was increased. Uteroplacental glutamine output to the fetus and expression of genes regulating glutamine-glutamate metabolism were lower in PI-IUGR. Fetal glycine uptake was lower in PI-IUGR, with no differences in uteroplacental glycine or serine flux. These results suggest increased placental utilization of pyruvate from the fetus, without higher maternal glucose utilization, and lower fetoplacental amino acid shuttling during PI-IUGR. Mechanistically, AMP-activated protein kinase (AMPK) activation was higher and associated with thiobarbituric acid-reactive substances (TBARS) content, a marker of oxidative stress, and PDH activity in the PI-IUGR placenta, supporting a potential link between oxidative stress, AMPK, and pyruvate utilization. These differences in fetoplacental nutrient sensing and shuttling may represent adaptive strategies enabling the placenta to maintain oxidative metabolism.NEW & NOTEWORTHY These results suggest increased placental utilization of pyruvate from the fetus, without higher maternal glucose uptake, and lower amino acid shuttling in the placental insufficiency-induced intrauterine growth restriction (PI-IUGR) placenta. AMPK activation was associated with oxidative stress and PDH activity, supporting a putative link between oxidative stress, AMPK, and pyruvate utilization. These differences in fetoplacental nutrient sensing and shuttling may represent adaptive strategies enabling the placenta to maintain oxidative metabolism at the expense of fetal growth.


Asunto(s)
Insuficiencia Placentaria , Humanos , Embarazo , Femenino , Animales , Ovinos , Insuficiencia Placentaria/metabolismo , Placenta/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Glutamina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Feto/metabolismo , Glucosa/metabolismo , Ácido Láctico/metabolismo , Aminoácidos/metabolismo , Nutrientes , Glicina/metabolismo , Serina/metabolismo , Piruvatos/metabolismo , Oxígeno/metabolismo
10.
J Nutr ; 153(4): 958-969, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36775243

RESUMEN

BACKGROUND: Placental insufficiency negatively impacts fetal growth and body composition (BC), potentially affecting lifelong health. Placental insufficiency, detectable as an abnormal umbilical artery resistance index (UmA-RI) on Doppler ultrasonography, is highly prevalent in otherwise healthy South African pregnant women. Appropriate intervention reduces stillbirth and perinatal death, but research on long-term outcomes of surviving infants is lacking. OBJECTIVES: This study aimed to describe and compare anthropometry and BC during the first 2 y of life in a cohort of term-born infants with normal and abnormal prenatal UmA-RI. METHODS: Term-born infants (n = 81; n = 55 normal, n = 26 abnormal UmA-RI on third trimester Doppler screening) were followed up at 8-time points until age 2 y. Anthropometric measurements were taken, and FFM and FM were assessed by deuterium dilution. Age- and sex-specific z-scores were calculated for anthropometric indices, FM, FFM, FM index (FMI), and FFM index (FFMI) using appropriate reference data. Anthropometry and BC of infants with normal and abnormal UmA-RI were compared using an independent t-test or Mann-Whitney test. RESULTS: At most ages, group mean z-scores were <0 for length-for-age and FM and >0 for weight-for-length and FFM. Compared with infants with normal UmA-RI, infants with abnormal UmA-RI had significantly lower weight-for-age z-scores at birth (-0.77 ± 0.75 compared with -0.30 ± 1.10, P = 0.026), ages 10 wk to 9 mo (-0.4 ± 0.87 to -0.2 ± 1.12 compared with 0.3 ± 0.85 to 0.6 ± 1.09; P = 0.007-0.017) and 18 mo (-0.6 ± 0.82 compared with 0.1 ± 1.18; P = 0.037); length-for-age z-scores at ages ≤14 wk (-1.3 ± 1.25 to -0.9 ± 0.87 compared with -0.2 ± 1.04 to -0.1 ± 1.00; P = 0.004-0.021); and FFM-for-age z-scores at ages ≤9 mo (-0.1 ± 0.82 to 0.7 ± 0.71 compared with 0.7 ± 1.00 to 1.3 ± 0.85; P = 0.002-0.028). FFMI, percentage FFM, FM, percentage FM, and FMI showed no consistent significant differences. CONCLUSIONS: Infants with abnormal UmA-RI had lower weight-for-age and length-for-age z-scores, particularly at younger ages, with proportionally lower FFM but no consistent differences in percentage FFM and FFMI. These findings merit further investigation in larger cohorts.


Asunto(s)
Insuficiencia Placentaria , Masculino , Recién Nacido , Humanos , Lactante , Femenino , Embarazo , Niño , Preescolar , Índice de Masa Corporal , Insuficiencia Placentaria/metabolismo , Sudáfrica , Placenta , Composición Corporal , Antropometría , Tejido Adiposo/metabolismo
11.
Eur J Obstet Gynecol Reprod Biol ; 278: 22-28, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36108451

RESUMEN

Placental insufficiency affects about 10% of pregnancies and can lead to pre-eclampsia, fetal growth restriction, and preterm birth. Despite significant advances in early prediction and prevention of preterm pre-eclampsia with aspirin, the effects of prophylaxis on fetal growth restriction are less certain, and the rates of late-onset pre-eclampsia are not influenced by aspirin treatment. Pregnancies complicated by placental insufficiency are characterized by increased oxidative stress, and recent studies suggest that melatonin has antioxidant properties and contributes to maintaining placental homeostasis. We aimed to systematically review the available literature about melatonin in pregnancies complicated by placental insufficiency, specifically preeclampsia and fetal growth restriction, exploring three different aspects: 1) maternal melatonin levels; 2) expression and activity of melatonin placental receptors; 3) effects of maternal melatonin administration. PubMed (Medline) and Scopus were searched until December 2020. Identified studies were screened and assessed independently by two authors. Data were extracted and compiled in qualitative evidence synthesis. The circadian pattern of melatonin secretion seems to be altered in pregnancies complicated by placental insufficiency reflected by lower production of melatonin, with consequent lower systemic and placental concentrations and lower expression of melatonin receptors, thus reducing the local release of the indole and its autocrine function. Small intervention studies also suggest that treatment is safe and may lead to prolongation of pregnancy and better outcomes, but double-blind, randomized placebo-controlled trials are lacking.


Asunto(s)
Melatonina , Insuficiencia Placentaria , Preeclampsia , Nacimiento Prematuro , Recién Nacido , Embarazo , Femenino , Humanos , Insuficiencia Placentaria/tratamiento farmacológico , Insuficiencia Placentaria/metabolismo , Preeclampsia/metabolismo , Melatonina/uso terapéutico , Melatonina/metabolismo , Retardo del Crecimiento Fetal/tratamiento farmacológico , Retardo del Crecimiento Fetal/metabolismo , Placenta/metabolismo , Antioxidantes/uso terapéutico , Antioxidantes/metabolismo , Receptores de Melatonina/metabolismo , Receptores de Melatonina/uso terapéutico , Nacimiento Prematuro/tratamiento farmacológico , Aspirina/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
12.
Fetal Diagn Ther ; 49(5-6): 279-292, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35760055

RESUMEN

Placental insufficiency is associated with reduced oxygen and nutrient supply to the fetus, which may result in fetal growth restriction (FGR). In an attempt to cope with the hostile intrauterine environment, FGR fetuses respond through metabolic, endocrine, vascular, cardiac, behavioral, hematological, and immunological adaptive mechanisms. However, permanent sequelae may result from such adaptive mechanisms. In this review, we describe the mechanisms of fetal adaptation to the hostile intrauterine environment in FGR of uteroplacental origin and detail their pathophysiology and potential implications for the extrauterine life of the individual.


Asunto(s)
Insuficiencia Placentaria , Femenino , Retardo del Crecimiento Fetal , Feto , Humanos , Hipoxia/complicaciones , Placenta/irrigación sanguínea , Insuficiencia Placentaria/metabolismo , Embarazo
13.
FASEB J ; 36(7): e22388, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35639049

RESUMEN

Epidemiological and animal experimental studies suggest an association between gestational cholestasis and intrauterine growth restriction (IUGR). Here, we explored the mechanism through which gestational cholestasis induced IUGR. To establish gestational cholestasis model, pregnant mice were subcutaneously injected with 17α-Ethynylestradiol (E2) on gestational day 13 (GD13)-GD17. Some pregnant mice were intraperitoneally injected with 4µ8C on GD13-GD17. The results found that the apoptosis of trophoblast cells was elevated in placentas of mice with gestational cholestasis and in deoxycholic acid (DCA)-treated human trophoblast cell lines and primary mouse trophoblast cells. Correspondingly, the levels of placental cleaved caspase-3 and Bax were increased, while placental Bcl2 level was decreased in mice with gestational cholestasis and in DCA-treated trophoblast cells. Further analysis found that placental IRE1α pathway was activated in mice with gestational cholestasis and in DCA-treated trophoblast cells. Interestingly, 4µ8C, an IRE1α RNase inhibitor, significantly inhibited caspase-3 activity and apoptosis of trophoblast cells in vivo and in vitro. Importantly, 4µ8C rescued gestational cholestasis-induced placental insufficiency and IUGR. Furthermore, a case-control study demonstrated that placental IRE1α and caspase-3 pathways were activated in cholestasis cases. Our results provide evidence that gestational cholestasis induces placental insufficiency and IUGR may be via triggering IRE1α-mediated apoptosis of placental trophoblast cells.


Asunto(s)
Colestasis Intrahepática , Endorribonucleasas , Insuficiencia Placentaria , Proteínas Serina-Treonina Quinasas , Animales , Apoptosis , Estudios de Casos y Controles , Caspasa 3/metabolismo , Colestasis Intrahepática/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Femenino , Retardo del Crecimiento Fetal/metabolismo , Humanos , Ratones , Placenta/metabolismo , Insuficiencia Placentaria/metabolismo , Embarazo , Complicaciones del Embarazo , Proteínas Serina-Treonina Quinasas/genética , Trofoblastos/metabolismo
14.
Placenta ; 125: 4-9, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35414477

RESUMEN

Pregnancy complications adversely impact both mother and/or fetus throughout the lifespan. Fetal growth restriction (FGR) occurs when a fetus fails to reach their intrauterine potential for growth, it is the second highest leading cause of infant mortality, and leads to increased risk of developing non-communicable diseases in later life due 'fetal programming'. Abnormal placental development, growth and/or function underlies approximately 75% of FGR cases and there is currently no treatment save delivery, often prematurely. We previously demonstrated in a murine model of FGR that nanoparticle mediated, intra-placental human IGF-1 gene therapy maintains normal fetal growth. Multiple models of FGR currently exist reflecting the etiologies of human FGR and have been used by us and others to investigate the development of in utero therapeutics as discussed here. In addition to the in vivo models discussed herein, utilizing human models including in vitro (Choriocarcinoma cell lines and primary trophoblasts) and ex vivo (term villous fragments and placenta cotyledon perfusion) we have demonstrated robust nanoparticle uptake, transgene expression, nutrient transporter regulation without transfer to the fetus. For translational gene therapy application in the human placenta, there are multiple avenues that require investigation including syncytial uptake from the maternal circulation, transgene expression, functionality and longevity of treatment, impact of treatment on the mother and developing fetus. The potential impact of treating the placenta during gestation is high, wide-ranging across pregnancy complications, and may offer reduced risk of developing associated cardio-metabolic diseases in later life impacting at both an individual and societal level.


Asunto(s)
Insuficiencia Placentaria , Animales , Femenino , Desarrollo Fetal , Retardo del Crecimiento Fetal/metabolismo , Humanos , Ratones , Placenta/metabolismo , Insuficiencia Placentaria/metabolismo , Placentación , Embarazo
15.
Reprod Sci ; 29(6): 1776-1789, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34611848

RESUMEN

Pregnant sheep have been used to model complications of human pregnancies including placental insufficiency and intrauterine growth restriction. Some of the hallmarks of placental insufficiency are slower uterine and umbilical blood flow rates, impaired placental transport of oxygen and amino acids, and lower fetal arterial concentrations of anabolic growth factors. An impact of fetal sex on these outcomes has not been identified in either human or sheep pregnancies. This is likely because most studies measuring these outcomes have used small numbers of subjects or animals. We undertook a secondary analysis of previously published data generated by our laboratory in late-gestation (gestational age of 133 ± 0 days gestational age) control sheep (n = 29 male fetuses; n = 26 female fetuses; n = 3 sex not recorded) and sheep exposed to elevated ambient temperatures to cause experimental placental insufficiency (n = 23 male fetuses; n = 17 female fetuses; n = 1 sex not recorded). The primary goal was to determine how fetal sex modifies the effect of the experimental insult on outcomes related to placental blood flow, amino acid and oxygen transport, and fetal hormones. Of the 112 outcomes measured, we only found an interaction between fetal sex and experimental insult for the uterine uptake rates of isoleucine, phenylalanine, and arginine. Additionally, most outcomes measured did not show a difference based on fetal sex when adjusting for the impact of placental insufficiency. Exceptions included fetal norepinephrine and cortisol concentrations, which were higher in female compared to male fetuses. For the parameters measured in the current analysis, the impact of fetal sex was not widespread.


Asunto(s)
Insuficiencia Placentaria , Aminoácidos/metabolismo , Aminoácidos/farmacología , Animales , Femenino , Retardo del Crecimiento Fetal/metabolismo , Feto/metabolismo , Humanos , Masculino , Oxígeno , Placenta/metabolismo , Circulación Placentaria , Insuficiencia Placentaria/metabolismo , Embarazo , Ovinos
16.
Am J Physiol Regul Integr Comp Physiol ; 322(3): R228-R240, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34907787

RESUMEN

Skeletal muscle from the late gestation sheep fetus with intrauterine growth restriction (IUGR) has evidence of reduced oxidative metabolism. Using a sheep model of placental insufficiency and IUGR, we tested the hypothesis that by late gestation, IUGR fetal skeletal muscle has reduced capacity for oxidative phosphorylation because of intrinsic deficits in mitochondrial respiration. We measured mitochondrial respiration in permeabilized muscle fibers from biceps femoris (BF) and soleus (SOL) from control and IUGR fetal sheep. Using muscles including BF, SOL, tibialis anterior (TA), and flexor digitorum superficialis (FDS), we measured citrate synthase (CS) activity, mitochondrial complex subunit abundance, fiber type distribution, and gene expression of regulators of mitochondrial biosynthesis. Ex vivo mitochondrial respiration was similar in control and IUGR muscle. However, CS activity was lower in IUGR BF and TA, indicating lower mitochondrial content, and protein expression of individual mitochondrial complex subunits was lower in IUGR TA and BF in a muscle-specific pattern. IUGR TA, BF, and FDS also had lower expression of type I oxidative fibers. Fiber-type shifts that support glycolytic instead of oxidative metabolism may be advantageous for the IUGR fetus in a hypoxic and nutrient-deficient environment, whereas these adaptions may be maladaptive in postnatal life.


Asunto(s)
Citrato (si)-Sintasa/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Estrés Oxidativo/fisiología , Animales , Femenino , Feto/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fosforilación Oxidativa , Placenta/metabolismo , Insuficiencia Placentaria/metabolismo , Embarazo , Ovinos
17.
Front Endocrinol (Lausanne) ; 12: 612888, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079518

RESUMEN

Intrauterine growth restriction (IUGR) of the fetus, resulting from placental insufficiency (PI), is characterized by low fetal oxygen and nutrient concentrations that stunt growth rates of metabolic organs. Numerous animal models of IUGR recapitulate pathophysiological conditions found in human fetuses with IUGR. These models provide insight into metabolic dysfunction in skeletal muscle and liver. For example, cellular energy production and metabolic rate are decreased in the skeletal muscle and liver of IUGR fetuses. These metabolic adaptations demonstrate that fundamental processes in mitochondria, such as substrate utilization and oxidative phosphorylation, are tempered in response to low oxygen and nutrient availability. As a central metabolic organelle, mitochondria coordinate cellular metabolism by coupling oxygen consumption to substrate utilization in concert with tissue energy demand and accretion. In IUGR fetuses, reducing mitochondrial metabolic capacity in response to nutrient restriction is advantageous to ensure fetal survival. If permanent, however, these adaptations may predispose IUGR fetuses toward metabolic diseases throughout life. Furthermore, these mitochondrial defects may underscore developmental programming that results in the sequela of metabolic pathologies. In this review, we examine how reduced nutrient availability in IUGR fetuses impacts skeletal muscle and liver substrate catabolism, and discuss how enzymatic processes governing mitochondrial function, such as the tricarboxylic acid cycle and electron transport chain, are regulated. Understanding how deficiencies in oxygen and substrate metabolism in response to placental restriction regulate skeletal muscle and liver metabolism is essential given the importance of these tissues in the development of later lifer metabolic dysfunction.


Asunto(s)
Retardo del Crecimiento Fetal/etiología , Mitocondrias/fisiología , Enfermedades Mitocondriales/complicaciones , Animales , Ciclo del Ácido Cítrico/fisiología , Femenino , Retardo del Crecimiento Fetal/metabolismo , Humanos , Recién Nacido , Hígado/metabolismo , Hígado/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fosforilación Oxidativa , Oxígeno/metabolismo , Insuficiencia Placentaria/etiología , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/patología , Embarazo
18.
Placenta ; 110: 1-8, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34051643

RESUMEN

INTRODUCTION: In this study, we aimed at quantifying placental concentrations of 22 chemical elements in small fetuses (SGA) as compared with normally grown fetuses (AGA), and to assess the relationship with Doppler markers of placental function. METHODS: Prospective cohort study, including 71 SGA fetuses (estimated fetal weight < 10th percentile) and 96 AGA fetuses (estimated fetal weight > 10th percentile), recruited in the third trimester of gestation. The placental concentration of 22 chemical elements was determined by inductively coupled plasma optical emission spectrophotometer (ICP-OES, ICAP 6500 Duo Thermo): aluminum (Al), beryllium (Be), bismuth (Bi), calcium (Ca), cadmium (Cd), cobalt (Co), chrome (Cr), copper (Cu), magnesium (Mg), manganese (Mn), molybdenum (Mo), nickel (Ni), phosphorus (P), lead (Pb), rubidium (Rb), sulfur (S), strontium (Sr), titanium (Ti), thallium (Tl), antimony (Sb), selenium (Se), and zinc (Zn). Placental function was assessed by measuring the following fetal-maternal parameters: Uterine artery Pulsatility Index (UtA PI), Umbilical artery Pulsatility Index (UA PI) and Middle Cerebral artery Pulsatility Index (MCA PI). The association between the chemical elements concentration and study group and the association with Doppler measures were evaluated. RESULTS: SGA was associated with significantly (p < 0.05) lower concentrations of Al (AGA 21.14 vs SGA 0.51 mg/kg), Cr (AGA 0.17 vs SGA 0.12 mg/kg), Cu (AGA 0.89 vs SGA 0.81 mg/kg), Mg (AGA 0.007 vs SGA 0.006 g/100g), Mn (AGA 0.60 vs SGA 0.47 mg/kg), Rb (AGA 1.68 vs SGA 1.47 mg/kg), Se (AGA 0.02 vs SGA 0.01 mg/kg), Ti (AGA 0.75 vs SGA 0.05 mg/kg) and Zn (AGA 9.04 vs SGA 8.22 mg/kg). Lower placental concentrations of Al, Cr, Mn, Se, Ti were associated with abnormal UtA, UA and MCA Doppler. DISCUSSION: Lower placental concentrations of Al, Cr, Cu, Mn, Rb, Se, Ti and Zn are associated with SGA fetuses and abnormal fetal-maternal Doppler results. Additional studies are required to further understand how chemical elements affect fetal growth and potentially find strategies to prevent SGA.


Asunto(s)
Elementos Químicos , Retardo del Crecimiento Fetal , Placenta/química , Placenta/fisiopatología , Adulto , Estudios de Cohortes , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Peso Fetal , Feto/diagnóstico por imagen , Edad Gestacional , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional , Masculino , Intercambio Materno-Fetal/fisiología , Placenta/diagnóstico por imagen , Placenta/metabolismo , Insuficiencia Placentaria/diagnóstico por imagen , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/patología , Insuficiencia Placentaria/fisiopatología , Embarazo , Estudios Prospectivos , Espectrofotometría Atómica , Ultrasonografía Doppler , Ultrasonografía Prenatal
19.
J Endocrinol ; 249(3): 195-207, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33994373

RESUMEN

Fetuses with intrauterine growth restriction (IUGR) have high concentrations of catecholamines, which lowers the insulin secretion and glucose uptake. Here, we studied the effect of hypercatecholaminemia on glucose metabolism in sheep fetuses with placental insufficiency-induced IUGR. Norepinephrine concentrations are elevated throughout late gestation in IUGR fetuses but not in IUGR fetuses with a bilateral adrenal demedullation (IAD) at 0.65 of gestation. Euglycemic (EC) and hyperinsulinemic-euglycemic (HEC) clamps were performed in control, intact-IUGR, and IAD fetuses at 0.87 of gestation. Compared to controls, basal oxygen, glucose, and insulin concentrations were lower in IUGR groups. Norepinephrine concentrations were five-fold higher in IUGR fetuses than in IAD fetuses. During the EC, rates of glucose entry (GER, umbilical + exogenous), glucose utilization (GUR), and glucose oxidation (GOR) were greater in IUGR groups than in controls. In IUGR and IAD fetuses with euglycemia and euinsulinemia, glucose production rates (GPR) remained elevated. During the HEC, GER and GOR were not different among groups. In IUGR and IAD fetuses, GURs were 40% greater than in controls, which paralleled the sustained GPR despite hyperinsulinemia. Glucose-stimulated insulin concentrations were augmented in IAD fetuses compared to IUGR fetuses. Fetal weights were not different between IUGR groups but were less than controls. Regardless of norepinephrine concentrations, IUGR fetuses not only develop greater peripheral insulin sensitivity for glucose utilization but also develop hepatic insulin resistance because GPR was maintained and unaffected by euglycemia or hyperinsulinemia. These findings show that adaptation in glucose metabolism of IUGR fetuses are independent of catecholamines, which implicate that hypoxemia and hypoglycemia cause the metabolic responses.


Asunto(s)
Catecolaminas/metabolismo , Retardo del Crecimiento Fetal/veterinaria , Glucosa/metabolismo , Norepinefrina/metabolismo , Glándulas Suprarrenales/patología , Animales , Transporte Biológico , Glucemia , Catecolaminas/sangre , Femenino , Desarrollo Fetal , Feto , Norepinefrina/sangre , Insuficiencia Placentaria/metabolismo , Embarazo , Ovinos
20.
Am J Obstet Gynecol ; 225(5): 520.e1-520.e10, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33901486

RESUMEN

BACKGROUND: The physiological control of human parturition at term is unknown. OBJECTIVE: This study aimed to test the hypothesis that slowing of fetal growth or elevated maternal serum levels of markers of placental hypoxia in late gestation will be associated with earlier term labor. STUDY DESIGN: We observed 2208 women having first births and performed serial blinded ultrasonography and immunoassay of soluble fms-like tyrosine kinase-1 and placenta growth factor. We estimated the probability of spontaneous delivery from 37 weeks of gestational age concerning (1) fetal growth between 20 and 36 weeks of gestational age and (2) the maternal serum soluble fms-like tyrosine kinase-1-to-placenta growth factor ratio measured at approximately 36 weeks of gestational age. Data were analyzed using logistic regression and Cox regression. RESULTS: Fetal size at 36 weeks of gestational age was not independently associated with the timing of delivery at term. However, there was an inverse relationship between fetal growth between 20 weeks of gestational age and 36 weeks of gestational age and the probability of spontaneous labor at 37 to 38 weeks' gestation (hazard ratio [95% confidence interval] for a 50 percentile increase in abdominal circumference growth velocity, 0.60 [0.47-0.78]; P=.0001). This association was weaker at 39 to 40 weeks' gestation (0.83 [0.74-0.93]; P=.0013), and there was no association at ≥41 weeks' gestation. Very similar associations were observed for estimated fetal weight growth velocity. There was a positive relationship between soluble fms-like tyrosine kinase-1-to-placenta growth factor ratio and the probability of spontaneous labor at 37 to 38 weeks' gestation (hazard ratio [95% confidence interval] for a 50 percentile increase in soluble fms-like tyrosine kinase-1-to-placenta growth factor ratio, 3.05 [2.32-4.02]; P<.0001). This association was weaker at 39 to 40 weeks' gestation (1.46 [1.30-1.63]; P<.0001), and there was no association at ≥41 weeks' gestation. Adjustment for maternal characteristics was without material effect on any of these associations. CONCLUSION: Slowing of fetal growth and biomarkers of placental insufficiency were associated with an increased probability of early onset of spontaneous term labor. We speculated that progressive placental insufficiency may be a physiological phenomenon that occurs with advancing gestational age near and at term and promotes the initiation of labor.


Asunto(s)
Desarrollo Fetal , Trabajo de Parto , Factor de Crecimiento Placentario/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Edad Gestacional , Humanos , Insuficiencia Placentaria/metabolismo , Embarazo , Ultrasonografía Prenatal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...