Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.909
Filtrar
Más filtros

Intervalo de año de publicación
1.
Hum Reprod ; 39(9): 1925-1933, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39025484

RESUMEN

STUDY QUESTION: Is periconceptional multiple-micronutrient supplement (MMS) use including folic acid (FA) compared to FA use only associated with increased embryonic growth, development, and birth weight in a high-risk population? SUMMARY ANSWER: Women with MMS intake show no significant differences in first-trimester morphological embryo development, but increased first-trimester embryonic growth trajectories and fewer neonates born small for gestational age (SGA), less than the 3rd percentile (

Asunto(s)
Suplementos Dietéticos , Desarrollo Embrionario , Ácido Fólico , Recién Nacido Pequeño para la Edad Gestacional , Humanos , Femenino , Embarazo , Ácido Fólico/administración & dosificación , Adulto , Desarrollo Embrionario/efectos de los fármacos , Recién Nacido , Estudios Prospectivos , Peso al Nacer/efectos de los fármacos , Países Bajos/epidemiología , Resultado del Embarazo , Atención Preconceptiva , Desarrollo Fetal/efectos de los fármacos , Estudios de Cohortes , Ultrasonografía Prenatal , Primer Trimestre del Embarazo
2.
Reproduction ; 168(3)2024 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-39096934

RESUMEN

In Brief: Aging in men is associated with diminished sperm quality and a higher incidence of altered fetal development and miscarriage in resultant pregnancies. This study in mice identifies a therapeutic compound that, when administered to aged males, improves sperm quality, subsequent embryo development and post-natal offspring health. Abstract: Aging in men is associated with diminished sperm quality and a higher incidence of altered fetal development and miscarriage in resultant pregnancies. We used a mouse model of advanced paternal age to characterize embryonic development in older male mice and tested whether pre-conception treatment with the mitochondrial activator BGP-15 improves reproductive outcomes in old males. Like older men, reproductively old male mice had higher levels of sperm DNA damage and delayed pre-implantation development, associated with a reduced fetal weight and placental weight. Analysis of neonatal outcomes of in vivo-conceived offspring found that pups sired by old males were smaller, had delayed locomotor development, and increased mortality. BGP-15 treatment for 5 days prior to conception reduced sperm DNA oxidation levels and improved on-time embryo development after IVF and pup survival. BGP-15 treatment for 3 weeks prior to conception improved on-time pre-implantation embryo development and fetal viability and increased fetal size in pregnancies sired by old males. These results validate that ageing negatively affects male fertility and offspring physiology and indicates that pre-conception treatment with BGP-15 has the potential to improve sperm quality as well as early embryo development and post-natal health.


Asunto(s)
Envejecimiento , Fertilidad , Espermatozoides , Animales , Masculino , Ratones , Espermatozoides/efectos de los fármacos , Femenino , Fertilidad/efectos de los fármacos , Embarazo , Desarrollo Embrionario/efectos de los fármacos , Reproducción/efectos de los fármacos , Ratones Endogámicos C57BL , Daño del ADN , Análisis de Semen , Desarrollo Fetal/efectos de los fármacos
3.
Crit Rev Toxicol ; 54(7): 431-464, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39016688

RESUMEN

During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.


Asunto(s)
Placenta , Humanos , Embarazo , Femenino , Placenta/efectos de los fármacos , Placentación/efectos de los fármacos , Modelos Biológicos , Desarrollo Fetal/efectos de los fármacos
4.
Crit Rev Toxicol ; 54(9): 619-633, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39093553

RESUMEN

Embryofetal development (EFD) studies are performed to characterize risk of drugs in pregnant women and on embryofetal development. In line with the ICH S5(R3) guideline, these studies are generally conducted in one rodent and one non-rodent species, commonly rats and rabbits. However, the added value of conducting EFD studies in two species to risk assessment is debatable. In this study, rat and rabbit EFD studies were evaluated to analyze the added value of a second species. Information on rat and rabbit EFD studies conducted for human pharmaceuticals submitted for marketing authorization to the European Medicines Agency between 2004 and 2022 was collected from the database of the Dutch Medicines Evaluation Board, along with EFD studies conducted for known human teratogens. In total, 369 compounds were included in the database. For 55.6% of the compounds similar effects were observed in rat and rabbit EFD studies. Discordance was observed for 44.6% of compounds. Discordance could often be explained based on occurrence of maternal toxicity or the compound's mechanism of action. For other compounds, discordance was considered of limited clinical relevance due to high exposure margins or less concerning EFD toxicity. For 6.2%, discordance could not be explained and was considered clinically relevant. Furthermore, for specific therapeutic classes, concordance between rat and rabbit could vary. In conclusion, in many cases the added value of conducting EFD studies in two species is limited. These data could help identify scenarios in which (additional) EFD studies could be waived or create a weight-of-evidence model to determine the need for (additional) EFD studies.


Asunto(s)
Desarrollo Embrionario , Teratógenos , Animales , Conejos , Ratas , Embarazo , Femenino , Desarrollo Embrionario/efectos de los fármacos , Teratógenos/toxicidad , Medición de Riesgo , Humanos , Pruebas de Toxicidad , Desarrollo Fetal/efectos de los fármacos , Especificidad de la Especie
5.
Arch Toxicol ; 98(6): 1891-1908, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38522057

RESUMEN

Dexamethasone is widely used in pregnant women at risk of preterm birth to reduce the occurrence of neonatal respiratory distress syndrome and subsequently reduce neonatal mortality. Studies have suggested that dexamethasone has developmental toxicity, but there is a notable absence of systematic investigations about its characteristics. In this study, we examined the effects of prenatal dexamethasone exposure (PDE) on mother/fetal mice at different doses (0.2, 0.4, or 0.8 mg/kg b.i.d), stages (gestational day 14-15 or 16-17) and courses (single- or double-course) based on the clinical practice. Results showed that PDE increased intrauterine growth retardation rate, and disordered the serum glucose, lipid and cholesterol metabolic phenotypes, and sex hormone level of mother/fetal mice. PDE was further discovered to interfere with the development of fetal lung, hippocampus and bone, inhibits steroid synthesis in adrenal and testis, and promotes steroid synthesis in the ovary and lipid synthesis in the liver, with significant effects observed at high dose, early stage and double course. The order of severity might be: ovary > lung > hippocampus/bone > others. Correlation analysis revealed that the decreased serum corticosterone and insulin-like growth factor 1 (IGF1) levels were closely related to PDE-induced low birth weight and abnormal multi-organ development in offspring. In conclusion, this study systematically confirmed PDE-induced multi-organ developmental toxicity, elucidated its characteristics, and proposed the potential "glucocorticoid (GC)-IGF1" axis programming mechanism. This research provided an experimental foundation for a comprehensive understanding of the effect and characteristics of dexamethasone on fetal multi-organ development, thereby guiding the application of "precision medicine" during pregnancy.


Asunto(s)
Dexametasona , Relación Dosis-Respuesta a Droga , Desarrollo Fetal , Animales , Femenino , Embarazo , Dexametasona/toxicidad , Dexametasona/administración & dosificación , Masculino , Desarrollo Fetal/efectos de los fármacos , Ratones , Retardo del Crecimiento Fetal/inducido químicamente , Factor I del Crecimiento Similar a la Insulina/metabolismo , Glucocorticoides/toxicidad , Glucocorticoides/administración & dosificación , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
6.
Arch Toxicol ; 98(7): 1975-1990, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38581585

RESUMEN

New human life begins in the uterus in a period of both extreme plasticity and sensitivity to environmental disturbances. The fetal stage is also a vital period for central nervous system development, with experiences at this point profoundly and permanently shaping brain structure and function. As such, some brain disorders may originate in utero. Glucocorticoids, a class of essential stress hormones, play indispensable roles in fetal development, but overexposure may have lasting impacts on the brain. In this review, we summarize data from recent clinical and non-clinical studies regarding alterations in fetal brains due to prenatal glucocorticoid overexposure that are associated with nervous system disorders. We discuss relevant changes to brain structure and cellular functions and explore the underlying molecular mechanisms. In addition, we summarize factors that may cause differential outcomes between varying brain regions, and outline clinically feasible intervention strategies that are expected to minimize negative consequences arising from fetal glucocorticoid overexposure. Finally, we highlight the need for experimental evidence aided by new technologies to clearly determine the effects of excessive prenatal glucocorticoid exposure. This review consolidates diverse findings to help researchers better understand the relationship between the prenatal glucocorticoid overexposure and the effects it has on various fetal brain regions, promoting further development of critical intervention strategies.


Asunto(s)
Encefalopatías , Encéfalo , Glucocorticoides , Efectos Tardíos de la Exposición Prenatal , Humanos , Glucocorticoides/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Femenino , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Animales , Encefalopatías/inducido químicamente , Desarrollo Fetal/efectos de los fármacos
7.
Regul Toxicol Pharmacol ; 152: 105684, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39151719

RESUMEN

Allyl alcohol (C3H6O; prop-2-en-1-ol; CAS RN 107-18-6; EINECS 203-470-7) is used as an intermediate/monomer in polymerization reactions producing chemicals/optical resins or as a coupling/cross-linking agent for unsaturated polyester and alkyd resins. Human exposure to allyl alcohol (AA) is restricted to workplace manufacturing facilities where it is used in enclosed systems, which limits release and impact on environmental receptors. To address regulatory questions about possible developmental toxicity, two OECD Guideline studies were conducted. A rat developmental toxicity study found fetal and maternal toxicity, in the form of resorptions and decreased body weight and food consumption, but no teratogenic effects. A rabbit developmental toxicity study was subsequently conducted upon request by the European Chemical Agency in 2011 under the REACH program and likewise reported maternal toxicity in the form of reductions in body weight gain and food consumption, but neither fetal toxicity or teratogenic effects. The results of both studies are presented and compared in this paper. Based on our review of the collective results of these studies, AA is considered non-teratogenic, yet does elicit increased post-implantation loss and reduced fetal body weight, possibly resulting from concomitant maternal toxicity. Based on the results of these studies, a maternal and developmental toxicity No Observed Adverse Effect Level of 10 mg/kg/day was apparent for both species.


Asunto(s)
Nivel sin Efectos Adversos Observados , Propanoles , Animales , Femenino , Conejos , Ratas , Embarazo , Propanoles/toxicidad , Desarrollo Fetal/efectos de los fármacos , Masculino , Pruebas de Toxicidad/métodos , Exposición Materna/efectos adversos
8.
Dev Psychobiol ; 66(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38646069

RESUMEN

Choline and folate are critical nutrients for fetal brain development, but the timing of their influence during gestation has not been previously characterized. At different periods during gestation, choline stimulation of α7-nicotinic receptors facilitates conversion of γ-aminobutyric acid (GABA) receptors from excitatory to inhibitory and recruitment of GluR1-R2 receptors for faster excitatory responses to glutamate. The outcome of the fetal development of inhibition and excitation was assessed in 159 newborns by P50 cerebral auditory-evoked responses. Paired stimuli, S1, S2, were presented 500 msec apart. Higher P50 amplitude in response to S1 (P50S1microV) assesses excitation, and lower P50S2microV assesses inhibition in this paired-stimulus paradigm. Development of inhibition was related solely to maternal choline plasma concentration and folate supplementation at 16 weeks' gestation. Development of excitation was related only to maternal choline at 28 weeks. Higher maternal choline concentrations later in gestation did not compensate for earlier lower concentrations. At 4 years of age, increased behavior problems on the Child Behavior Checklist 1½-5yrs were related to both newborn inhibition and excitation. Incomplete development of inhibition and excitation associated with lower choline and folate during relatively brief periods of gestation thus has enduring effects on child development.


Asunto(s)
Colina , Potenciales Evocados Auditivos , Ácido Fólico , Humanos , Colina/farmacología , Colina/metabolismo , Femenino , Ácido Fólico/farmacología , Masculino , Recién Nacido , Embarazo , Potenciales Evocados Auditivos/fisiología , Potenciales Evocados Auditivos/efectos de los fármacos , Preescolar , Desarrollo Fetal/fisiología , Desarrollo Fetal/efectos de los fármacos , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de los fármacos , Adulto , Edad Gestacional , Desarrollo Infantil/fisiología , Desarrollo Infantil/efectos de los fármacos
9.
Ecotoxicol Environ Saf ; 283: 116980, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39226632

RESUMEN

OBJECTIVE: Acetaminophen (APAP), an antipyretic and analgesic commonly used during pregnancy, has been recognized as a novel environmental contaminant. Preliminary evidence suggests that prenatal acetaminophen exposure (PAcE) could adversely affect offspring's gonadal and neurologic development, but there is no systematic investigation on the characteristics of APAP's fetal developmental toxicity. METHODS: Pregnant mice were treated with 100 or 400 mg/kg∙d APAP in the second-trimester, or 400 mg/kg∙d APAP in the second- or third-trimester, or different courses (single or multiple) of APAP, based on clinical regimen. The effects of PAcE on pregnancy outcomes, maternal/fetal blood phenotypes, and multi-organ morphological and functional development of fetal mice were analyzed. RESULTS: PAcE increased the incidence of adverse pregnancy outcomes and altered blood phenotypes including aminotransferases, lipids, and sex hormones in dams and fetuses. The expression of key functional genes in fetal organs indicated that PAcE inhibited hippocampal synaptic development, sex hormone synthesis, and osteogenic and chondrogenic development, but enhanced hepatic lipid synthesis and uptake, renal inflammatory hyperplasia, and adrenal steroid hormone synthesis. PAcE also induced marked pathological alterations in the fetal hippocampus, bone, kidney, and cartilage. The sensitivity rankings of fetal organs to PAcE might be hippocampus/bone > kidney > cartilage > liver > gonad > adrenal gland. Notably, PAcE-induced multi-organ developmental toxicity was more considerable under high-dose, second-trimester, and multi-course exposure and in male fetuses. CONCLUSION: This study confirmed PAcE-induced alterations in multi-organ development and function in fetal mice and elucidated its characteristics, which deepens the comprehensive understanding of APAP's developmental toxicity.


Asunto(s)
Acetaminofén , Animales , Acetaminofén/toxicidad , Femenino , Embarazo , Ratones , Masculino , Desarrollo Fetal/efectos de los fármacos , Analgésicos no Narcóticos/toxicidad , Exposición Materna , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Feto/efectos de los fármacos , Resultado del Embarazo
10.
Ecotoxicol Environ Saf ; 280: 116526, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38823346

RESUMEN

OBJECTIVES: Fetal overgrowth has detrimental effects on both the mother and the fetus. The global issue of ambient air pollution has been found to contribute to fetal overgrowth through various pathways. This study aimed to identify the association between prenatal exposure to ambient air pollution and the risk of fetal overgrowth. METHODS: We identified articles between January 2013 and February 2024 by searching the Web of Sciences(WoS), PubMed, Proquest, Scopus, and Google Scholar databases. Quality assessment was performed using the Newcastle Ottawa scale. This review was provided based on the PRISMA guideline and registered with PROSPERO, "CRD42023488936". RESULTS: The search generated 1719 studies, of which 22 cohort studies were included involving 3,480,041 participants. Results on the effects of air pollutants on fetal overgrowth are inconsistent because they vary in population and geographic region. But in general, the results indicate that prenatal exposure to air pollutants, specifically PM2.5, NO2, and SO2, is linked to a higher likelihood of fetal overgrowth(macrosomia and large for gestational age). Nevertheless, the relationship between CO and O3 pollution and fetal overgrowth remains uncertain. Furthermore, PM10 has a limited effect on fetal overgrowth. It is essential to consider the time that reproductive-age women are exposed to air pollution. Exposure to air pollutants before conception and throughout pregnancy has a substantial impact on the fetus's vulnerability to overgrowth. CONCLUSIONS: Fetal overgrowth has implications for the health of both mother and fetus. fetal overgrowth can cause cardiovascular diseases, obesity, type 2 diabetes, and other diseases in adulthood, so it is considered an important issue for the health of the future generation. Contrary to popular belief that air pollution leads to intrauterine growth restriction and low birth weight, this study highlights that one of the adverse consequences of air pollution is macrosomia or LGA during pregnancy. Therefore governments must focus on implementing initiatives that aim to reduce pregnant women's exposure to ambient air pollution to ensure the health of future generations.


Asunto(s)
Contaminación del Aire , Macrosomía Fetal , Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Femenino , Humanos , Embarazo , Contaminación del Aire/efectos adversos , Estudios de Cohortes , Desarrollo Fetal/efectos de los fármacos , Exposición Materna/efectos adversos , Material Particulado , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Macrosomía Fetal/etiología
11.
An Acad Bras Cienc ; 96(3): e20230604, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39016352

RESUMEN

This study aims to evaluate the phytochemical properties of Bauhinia holophylla (Bong.) Steud leaf extract, and their impact on maternal reproductive and fetal development in diabetic rats. For this, adult female Wistar rats (100 days of life) received streptozotocin (40 mg/Kg, intraperitoneal) for induction of diabetes, were mated and distributed into four groups: Nondiabetic; Nondiabetic given B. holophylla; Diabetic; and Diabetic given B. holophylla. The plant extract was given by gavage at increasing doses: 200, 400, and 800 mg/Kg. At day 21 of pregnancy, liver and blood samples were obtained for oxidative parameters and biochemical analysis, respectively. The uterus was removed for maternal-fetal outcomes. Phytochemical analysis showed a high content of phenolic components and biogenic amines. B. holophylla extract did not alter the glycemic levels but improved the lipid profile in diabetic animals. Besides that, the number of live fetuses and maternal weight gain were decreased in Diabetic group, and were not observed in animals treated. The group Diabetic treated presented a higher percentage of fetuses classified as adequate for gestational age compared to the Diabetic group. However, the treatment with plant extract caused embryo losses, fetal growth restriction, and teratogenicity in nondiabetic rats. Thus, the indiscriminate consumption requires carefulness.


Asunto(s)
Bauhinia , Diabetes Mellitus Experimental , Hipoglucemiantes , Extractos Vegetales , Ratas Wistar , Animales , Femenino , Extractos Vegetales/farmacología , Extractos Vegetales/química , Bauhinia/química , Embarazo , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Ratas , Fitoquímicos/farmacología , Fitoquímicos/análisis , Desarrollo Fetal/efectos de los fármacos , Estreptozocina , Glucemia/efectos de los fármacos , Glucemia/análisis , Hojas de la Planta/química
12.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731818

RESUMEN

Early life exposure lays the groundwork for the risk of developing cardiovascular-kidney-metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) has a key role as a ligand-activated transcription factor in sensing these environmental chemicals. Activating AHR through exposure to environmental chemicals has been documented for its adverse impacts on cardiovascular diseases, hypertension, diabetes, obesity, kidney disease, and non-alcoholic fatty liver disease, as evidenced by both epidemiological and animal studies. In this review, we compile current human evidence and findings from animal models that support the connection between antenatal chemical exposures and CKM programming, focusing particularly on AHR signaling. Additionally, we explore potential AHR modulators aimed at preventing CKM syndrome. As the pioneering review to present evidence advocating for the avoidance of toxic chemical exposure during pregnancy and deepening our understanding of AHR signaling, this has the potential to mitigate the global burden of CKM syndrome in the future.


Asunto(s)
Enfermedades Cardiovasculares , Efectos Tardíos de la Exposición Prenatal , Receptores de Hidrocarburo de Aril , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Humanos , Embarazo , Animales , Femenino , Efectos Tardíos de la Exposición Prenatal/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología , Exposición Materna/efectos adversos , Transducción de Señal/efectos de los fármacos , Riñón/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Desarrollo Fetal/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Contaminantes Ambientales/efectos adversos , Reprogramación Metabólica
13.
J Environ Sci (China) ; 145: 75-87, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38844325

RESUMEN

Prednisone is a synthetic glucocorticoid that is commonly used in both human and veterinary medication. Now, it is also recognized as an emerging environmental contaminant. Pregnant women may be exposed to prednisone actively or passively through multiple pathways and cause developmental toxicity to the fetus. However, the impact of prenatal prednisone exposure (PPE) on fetal kidney development remains unclear. In this study, pregnant mice were administered prednisone intragastrically during full-term pregnancy with different doses (0.25, 0.5, or 1 mg/(kg·day)), or at the dose of 1 mg/(kg·day) in different gestational days (GD) (GD0-9, GD10-18, or GD0-18). The pregnant mice were euthanized on GD18. HE staining revealed fetal kidney dysplasia, with an enlarged glomerular Bowman's capsule space and a reduced capillary network in the PPE groups. The expression of the podocyte and the mesangial cell marker genes was significantly reduced in the PPE groups. However, overall gene expression in renal tubules and collecting ducts were markedly increased. All of the above effects were more pronounced in high-dose, full-term pregnancy, and female fetuses. Studies on the mechanism of the female fetal kidney have revealed that PPE reduced the expression of Six2, increased the expression of Hnf1ß, Hnf4α, and Wnt9b, and inhibited the expression of glial cell line-derived neurotrophic factor (GDNF) and Notch signaling pathways. In conclusion, this study demonstrated that there is a sex difference in the developmental toxicity of PPE to the fetal kidney, and the time effect is manifested as full-term pregnancy > early pregnancy > mid-late pregnancy.


Asunto(s)
Riñón , Prednisona , Femenino , Animales , Embarazo , Ratones , Riñón/efectos de los fármacos , Riñón/embriología , Prednisona/toxicidad , Desarrollo Fetal/efectos de los fármacos , Masculino , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Exposición Materna/efectos adversos
14.
Proc Natl Acad Sci U S A ; 117(18): 10035-10044, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32312804

RESUMEN

One factor that contributes to the high prevalence of fetal alcohol spectrum disorder (FASD) is binge-like consumption of alcohol before pregnancy awareness. It is known that treatments are more effective with early recognition of FASD. Recent advances in retrospective motion correction for the reconstruction of three-dimensional (3D) fetal brain MRI have led to significant improvements in the quality and resolution of anatomical and diffusion MRI of the fetal brain. Here, a rhesus macaque model of FASD, involving oral self-administration of 1.5 g/kg ethanol per day beginning prior to pregnancy and extending through the first 60 d of a 168-d gestational term, was utilized to determine whether fetal MRI could detect alcohol-induced abnormalities in brain development. This approach revealed differences between ethanol-exposed and control fetuses at gestation day 135 (G135), but not G110 or G85. At G135, ethanol-exposed fetuses had reduced brainstem and cerebellum volume and water diffusion anisotropy in several white matter tracts, compared to controls. Ex vivo electrophysiological recordings performed on fetal brain tissue obtained immediately following MRI demonstrated that the structural abnormalities observed at G135 are of functional significance. Specifically, spontaneous excitatory postsynaptic current amplitudes measured from individual neurons in the primary somatosensory cortex and putamen strongly correlated with diffusion anisotropy in the white matter tracts that connect these structures. These findings demonstrate that exposure to ethanol early in gestation perturbs development of brain regions associated with motor control in a manner that is detectable with fetal MRI.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Encéfalo/fisiopatología , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Imagen de Difusión por Resonancia Magnética , Modelos Animales de Enfermedad , Etanol/toxicidad , Femenino , Trastornos del Espectro Alcohólico Fetal/diagnóstico por imagen , Desarrollo Fetal/efectos de los fármacos , Feto/diagnóstico por imagen , Feto/efectos de los fármacos , Humanos , Macaca mulatta , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/diagnóstico por imagen , Estudios Retrospectivos
15.
Drug Chem Toxicol ; 46(3): 609-615, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35502509

RESUMEN

Morinda citrifolia L., also known as Noni, is widely used plant in folk medicine for various therapeutic purposes. However, reports on its effects during pregnancy are limited. Therefore, the objective of this study was to evaluate the effects of the M. citrifolia fruit extract on maternal performance and fetal development during pregnancy in rats. Pregnant Wistar rats (n = 12/group) were treated from gestational days (GD) 0-21 with water (control group) or the aqueous extract of M. citrifolia fruit at doses of 200, 400, or 750 mg/kg, orally. During pregnancy, clinical signs of toxicity, maternal weight, feed intake, and water consumption were noted. On GD 21, the rats were anesthetized and blood was collected to evaluate various biochemical parameters. During laparotomy, reproductive performance parameters were recorded, and fetuses were weighed and the anomalies analyzed. Reduced placental efficiency and fetal growth restriction were observed in the group treated with 400 mg/kg of M. citrifolia extract. The highest dose (750 mg/kg) augmented aspartate aminotransferase concentration and preimplantation losses, while reducing the number of live fetuses. Furthermore, both doses (400 and 750 mg/kg) of the plant extract caused fetal anomalies. In conclusion, consumption of high doses of the M. citrifolia aqueous extrac during pregnancy leads to maternal hepatotoxicity, anti-implantation effects, intrauterine growth restriction and fetal abnormalities, indicating that the plant fruit extract can be harmful to both the mother and the fetus.


Asunto(s)
Desarrollo Fetal , Morinda , Placenta , Extractos Vegetales , Animales , Femenino , Embarazo , Ratas , Desarrollo Fetal/efectos de los fármacos , Frutas , Morinda/toxicidad , Placenta/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/toxicidad , Ratas Wistar
16.
Ceska Gynekol ; 88(3): 221-230, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37344189

RESUMEN

OBJECTIVE: Review of recent literature dealing with the effect of antipsychotic use during pregnancy on early postpartum adaptation of exposed infants and the development of congenital malformations. RESULTS: The use of antipsychotics during pregnancy does not appear to lead to significantly higher risk of congenital malformations but may pose a greater risk for the early adaptation of the newborn (especially the risk of preterm birth and intensive care unit admission). The study to date face methodological limitations - lack of information on exact doses of antipsychotics, lack of control groups of women with psychiatric problems but not taking antipsychotics and failure to control for confounding factors. CONCLUSION: The available data suggest the relative safety of antipsychotics during pregnancy, provided that potential risks are known, and the woman and her baby are carefully monitored.


Asunto(s)
Antipsicóticos , Desarrollo Fetal , Trastornos Mentales , Nacimiento Prematuro , Femenino , Humanos , Recién Nacido , Embarazo , Antipsicóticos/efectos adversos , Trastornos Mentales/tratamiento farmacológico , Complicaciones del Embarazo/psicología , Desarrollo Fetal/efectos de los fármacos
17.
Am J Obstet Gynecol ; 226(1): 130.e1-130.e11, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34364844

RESUMEN

BACKGROUND: Prenatal alcohol exposure is the most common cause of birth defects and intellectual disabilities and can increase the risk of stillbirth and negatively impact fetal growth. OBJECTIVE: To determine the effect of early prenatal alcohol exposure on nonhuman primate placental function and fetal growth. We hypothesized that early chronic prenatal alcohol would alter placental perfusion and oxygen availability that adversely affects fetal growth. STUDY DESIGN: Rhesus macaques self-administered 1.5 g/kg/d of ethanol (n=12) or isocaloric maltose-dextrin (n=12) daily before conception through the first 60 days of gestation (term is approximately 168 days). All animals were serially imaged with Doppler ultrasound to measure fetal biometry, uterine artery volume blood flow, and placental volume blood flow. Following Doppler ultrasound, all animals underwent both blood oxygenation level-dependent magnetic resonance imaging to characterize placental blood oxygenation and dynamic contrast-enhanced magnetic resonance imaging to quantify maternal placental perfusion. Animals were delivered by cesarean delivery for placental collection and fetal necropsy at gestational days 85 (n=8), 110 (n=8), or 135 (n=8). Histologic and RNA-sequencing analyses were performed on collected placental tissue. RESULTS: Placental volume blood flow was decreased at all gestational time points in ethanol-exposed vs control animals, but most significantly at gestational day 110 by Doppler ultrasound (P<.05). A significant decrease in total volumetric blood flow occurred in ethanol-exposed vs control animals on dynamic contrast-enhanced magnetic resonance imaging at both gestation days 110 and 135 (P<.05); moreover, a global reduction in T2∗, high blood deoxyhemoglobin concentration, occurred throughout gestation (P<.05). Similarly, evidence of placental ischemic injury was notable by histologic analysis, which revealed a significant increase in microscopic infarctions in ethanol-exposed, not control, animals, largely present at middle to late gestation. Fetal biometry and weight were decreased in ethanol-exposed vs control animals, but the decrease was not significant. Analysis with RNA sequencing suggested the involvement of the inflammatory and extracellular matrix response pathways. CONCLUSION: Early chronic prenatal alcohol exposure significantly diminished placental perfusion at mid to late gestation and also significantly decreased the oxygen supply to the fetal vasculature throughout pregnancy, these findings were associated with the presence of microscopic placental infarctions in the nonhuman primate. Although placental adaptations may compensate for early environmental perturbations to fetal growth, placental blood flow and oxygenation were reduced, consistent with the evidence of placental ischemic injury.


Asunto(s)
Etanol/efectos adversos , Macaca mulatta , Efectos Tardíos de la Exposición Prenatal/etiología , Animales , Modelos Animales de Enfermedad , Etanol/farmacología , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Placenta/efectos de los fármacos , Embarazo
18.
Regul Toxicol Pharmacol ; 129: 105119, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35031383

RESUMEN

The toxicological effects of DS-7309, a glucokinase activator, on pregnancy and embryo-fetal development in rats and rabbits and maternal blood glucose levels were examined. DS-7309 was administered at 3, 10, or 100 mg/kg to rats from Days 7-17 of pregnancy or at 10, 30, or 100 mg/kg to rabbits from Days 6-18 of pregnancy. In rats, maternal hypoglycemia (approximately 50 mg/dL) was seen at 3 and 10 mg/kg, but it recovered 7 h after dosing, leading to no toxic changes. In contrast, continuous severe maternal hypoglycemia (approximately 40 mg/dL, ≥7 h), fetal eye anomalies, and decreased fetal body weight were noted at 100 mg/kg. In rabbits, no fetal anomalies were seen at 10 and 30 mg/kg where maternal blood glucose level dropped to approximately 60-90 mg/dL, but recovered by 7 h after dosing at the latest. In contrast, at 100 mg/kg, severe hypoglycemia (around 60 mg/dL) was maintained and did not recover until 24 h after dosing; it resulted in decreased fetal viability and increased fetal skeleton anomalies. These findings indicate that DS-7309 could lead to embryo-fetal toxicity in rats and rabbits, with such toxicity considered to be related to continuous severe maternal hypoglycemia.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Hipoglucemiantes/farmacología , Animales , Área Bajo la Curva , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Glucoquinasa/metabolismo , Hipoglucemiantes/farmacocinética , Tasa de Depuración Metabólica , Conejos , Ratas , Ratas Sprague-Dawley
19.
Am J Physiol Endocrinol Metab ; 320(3): E527-E538, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427051

RESUMEN

Insulin-like growth factor-1 (IGF-1) is an important fetal growth factor. However, the role of fetal IGF-1 in increasing placental blood flow, nutrient transfer, and nutrient availability to support fetal growth and protein accretion is not well understood. Catheterized fetuses from late gestation pregnant sheep received an intravenous infusion of LR3 IGF-1 (LR3 IGF-1; n = 8) or saline (SAL; n = 8) for 1 wk. Sheep then underwent a metabolic study to measure uterine and umbilical blood flow, nutrient uptake rates, and fetal protein kinetic rates. By the end of the infusion, fetal weights were not statistically different between groups (SAL: 3.260 ± 0.211 kg, LR3 IGF-1: 3.682 ± 0.183; P = 0.15). Fetal heart, adrenal gland, and spleen weights were higher (P < 0.05), and insulin was lower in LR3 IGF-1 (P < 0.05). Uterine and umbilical blood flow and umbilical uptake rates of glucose, lactate, and oxygen were similar between groups. Umbilical amino acid uptake rates were lower in LR3 IGF-1 (P < 0.05) as were fetal concentrations of multiple amino acids. Fetal protein kinetic rates were similar. LR3 IGF-1 skeletal muscle had higher myoblast proliferation (P < 0.05). In summary, LR3 IGF-1 infusion for 1 wk into late gestation fetal sheep increased the weight of some fetal organs. However, because umbilical amino acid uptake rates and fetal plasma amino acid concentrations were lower in the LR3 IGF-1 group, we speculate that animals treated with LR3 IGF-1 can efficiently utilize available nutrients to support organ-specific growth in the fetus rather than by stimulating placental blood flow or nutrient transfer to the fetus.NEW & NOTEWORTHY After a 1-wk infusion of LR3 IGF-1, late gestation fetal sheep had lower umbilical uptake rates of amino acids, lower fetal arterial amino acid and insulin concentrations, and lower fetal oxygen content; however, LR-3 IGF-1-treated fetuses were still able to effectively utilize the available nutrients and oxygen to support organ growth and myoblast proliferation.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Nutrientes/metabolismo , Animales , Metabolismo Energético/efectos de los fármacos , Femenino , Sangre Fetal/metabolismo , Peso Fetal/efectos de los fármacos , Feto/efectos de los fármacos , Feto/metabolismo , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/embriología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Placenta/efectos de los fármacos , Placentación/efectos de los fármacos , Embarazo , Ovinos
20.
Toxicol Appl Pharmacol ; 426: 115638, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34242569

RESUMEN

Gonadal development begins in the intrauterine phase and females from most species are born with an established oocyte reserve. Exposure to drugs during gestation can compromise the offspring health, also affecting the gametes quality. Nicotine, the main component of cigarettes, is an oxidant agent capable of altering the fertility in men and women. As female gametes are susceptible to oxidative stress, this drug can damage the oolemma and affect oocyte maturation, induce errors during chromosomal segregation and DNA fragmentation. Oocyte mitochondria are particularly susceptible to injuries, contributing to the oocyte quality loss and embryonic development disruption. Thus, considering the high number of women who smoke during pregnancy, while significant events are occurring in the embryo for future fertility of offspring, we seek to verify the quality of the oocytes from adult rats exposed to nicotine during intrauterine phase and breastfeeding. Pregnant Wistar rats received nicotine by osmotic mini-pumps and the female progenies were evaluated in adulthood for oocyte quality (viability, lipid peroxidation, generation of reactive oxygen species and mitochondrial integrity) and reproductive capacity. Embryos (3dpc) and fetuses (20dpc) generated by these rats were also evaluated. The results showed that the dose of 2 mg/kg/day of nicotine through placenta and breast milk does not affect the number of oocytes and the fertility capacity of adult rats. However, it causes some morphological alterations in oocytes, mitochondrial changes, embryonic fragmentation and disruption of fetal development. The malformations in fetuses generated from these gametes can also indicate the occurrence of epigenetic modifications.


Asunto(s)
Nicotina/toxicidad , Oocitos/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Reproducción/efectos de los fármacos , Animales , Femenino , Desarrollo Fetal/efectos de los fármacos , Lactancia , Peroxidación de Lípido/efectos de los fármacos , Masculino , Intercambio Materno-Fetal , Mitocondrias/efectos de los fármacos , Oocitos/metabolismo , Embarazo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA