Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Physiol ; 602(9): 2127-2139, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38285002

RESUMEN

Maternal stress and glucocorticoid exposure during pregnancy have multigenerational effects on neuroendocrine function and behaviours in offspring. Importantly, effects are transmitted through the paternal lineage. Altered phenotypes are associated with profound differences in transcription and DNA methylation in the brain. In the present study, we hypothesized that maternal prenatal synthetic glucocorticoid (sGC) exposure in the F0 pregnancy will result in differences in miRNA levels in testes germ cells and sperm across multiple generations, and that these changes will associate with modified microRNA (miRNA) profiles and gene expression in the prefrontal cortex (PFC) of subsequent generations. Pregnant guinea-pigs (F0) were treated with multiple courses of the sGC betamethasone (Beta) (1 mg kg-1; gestational days 40, 41, 50, 51, 60 and 61) in late gestation. miRNA levels were assessed in testes germ cells and in F2 PFC using the GeneChip miRNA 4.0 Array and candidate miRNA measured in epididymal sperm by quantitative real-time PCR. Maternal Beta exposure did not alter miRNA levels in germ cells derived from the testes of adult male offspring. However, there were significant differences in the levels of four candidate miRNAs in the sperm of F1 and F2 adult males. There were no changes in miRNA levels in the PFC of juvenile F2 female offspring. The present study has identified that maternal Beta exposure leads to altered miRNA levels in sperm that are apparent for at least two generations. The fact that differences were confined to epididymal sperm suggests that the intergenerational effects of Beta may target the epididymis. KEY POINTS: Paternal glucocorticoid exposure prior to conception leads to profound epigenetic changes in the brain and somatic tissues in offspring, and microRNAs (miRNAs) in sperm may mediate these changes. We show that there were significant differences in the miRNA profile of epididymal sperm in two generations following prenatal glucocorticoid exposure that were not observed in germ cells derived from the testes. The epididymis is a probable target for intergenerational programming. The effects of prenatal glucocorticoid treatment may span multiple generations.


Asunto(s)
Glucocorticoides , MicroARNs , Efectos Tardíos de la Exposición Prenatal , Espermatozoides , Animales , Femenino , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Cobayas , Glucocorticoides/farmacología , Testículo/efectos de los fármacos , Testículo/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Betametasona/farmacología , Exposición Materna/efectos adversos
2.
Protein J ; 40(4): 466-488, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34110586

RESUMEN

The limb-girdle muscular dystrophies (LGMD) are a collection of genetic diseases united in their phenotypical expression of pelvic and shoulder area weakness and wasting. More than 30 subtypes have been identified, five dominant and 26 recessive. The increase in the characterization of new genotypes in the family of LGMDs further adds to the heterogeneity of the disease. Meanwhile, better understanding of the phenotype led to the reconsideration of the disease definition, which resulted in eight old subtypes to be no longer recognized officially as LGMD and five new diseases to be added to the LGMD family. The unique variabilities of LGMD stem from genetic mutations, which then lead to protein and ultimately muscle dysfunction. Herein, we review the LGMD pathway, starting with the genetic mutations that encode proteins involved in muscle maintenance and repair, and including the genotype-phenotype relationship of the disease, the epidemiology, disease progression, burden of illness, and emerging treatments.


Asunto(s)
Genotipo , Distrofia Muscular de Cinturas/clasificación , Distrofia Muscular de Cinturas/genética , Mutación , Humanos , Distrofia Muscular de Cinturas/epidemiología , Distrofia Muscular de Cinturas/terapia
3.
Sci Rep ; 9(1): 18211, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796763

RESUMEN

Synthetic glucocorticoids (sGC) are administered to women at risk for pre-term delivery, to mature the fetal lung and decrease neonatal morbidity. sGC also profoundly affect the fetal brain. The hippocampus expresses high levels of glucocorticoid (GR) and mineralocorticoid receptor (MR), and its development is affected by elevated fetal glucocorticoid levels. Antenatal sGC results in neuroendocrine and behavioral changes that persist in three generations of female guinea pig offspring of the paternal lineage. We hypothesized that antenatal sGC results in transgenerational changes in gene expression that correlate with changes in DNA methylation. We used RNASeq and capture probe bisulfite sequencing to investigate the transcriptomic and epigenomic effects of antenatal sGC exposure in the hippocampus of three generations of juvenile female offspring from the paternal lineage. Antenatal sGC exposure (F0 pregnancy) resulted in generation-specific changes in hippocampal gene transcription and DNA methylation. Significant changes in individual CpG methylation occurred in RNApol II binding regions of small non-coding RNA (snRNA) genes, which implicates alternative splicing as a mechanism involved in transgenerational transmission of the effects of antenatal sGC. This study provides novel perspectives on the mechanisms involved in transgenerational transmission and highlights the importance of human studies to determine the longer-term effects of antenatal sGC on hippocampal-related function.


Asunto(s)
Glucocorticoides/efectos adversos , Hipocampo/patología , Patrón de Herencia/efectos de los fármacos , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Femenino , Cobayas , Hipocampo/efectos de los fármacos , Humanos , Masculino , Embarazo , Nacimiento Prematuro/prevención & control , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/patología , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transcripción Genética/efectos de los fármacos
4.
Sci Rep ; 9(1): 764, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679753

RESUMEN

Synthetic glucocorticoids (sGC) are administered to women at risk for pre-term delivery to reduce respiratory distress syndrome in the newborn. The prefrontal cortex (PFC) is important in regulating stress responses and related behaviours and expresses high levels of glucocorticoid receptors (GR). Further, antenatal exposure to sGC results in a hyperactive phenotype in first generation (F1) juvenile male and female offspring, as well as F2 and F3 juvenile females from the paternal lineage. We hypothesized that multiple courses of antenatal sGC modify gene expression in the PFC, that these effects are sex-specific and maintained across multiple generations, and that the gene sets affected relate to modified locomotor activity. We performed RNA sequencing on PFC of F1 juvenile males and females, as well as F2 and F3 juvenile females from the paternal lineage and used regression modelling to relate gene expression and behavior. Antenatal sGC resulted in sex-specific and generation-specific changes in gene expression. Further, the expression of 4 genes (C9orf116, Calb1, Glra3, and Gpr52) explained 20-29% of the observed variability in locomotor activity. Antenatal exposure to sGC profoundly influences the developing PFC; effects are evident across multiple generations and may drive altered behavioural phenotypes.


Asunto(s)
Glucocorticoides/administración & dosificación , Locomoción/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Nacimiento Prematuro/tratamiento farmacológico , Síndrome de Dificultad Respiratoria del Recién Nacido/tratamiento farmacológico , Adulto , Calbindina 1/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucocorticoides/efectos adversos , Glucocorticoides/síntesis química , Humanos , Recién Nacido , Locomoción/genética , Masculino , Corteza Prefrontal/metabolismo , Nacimiento Prematuro/genética , Nacimiento Prematuro/patología , Nacimiento Prematuro/prevención & control , Receptores Acoplados a Proteínas G/genética , Receptores de Glicina/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Síndrome de Dificultad Respiratoria del Recién Nacido/prevención & control , Caracteres Sexuales , Estrés Fisiológico/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
5.
Endocrinology ; 159(12): 4065-4076, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30383219

RESUMEN

Treatment with a single course of synthetic glucocorticoids (sGCs) is the standard of care for pregnant women who are at risk for preterm delivery. Animal studies have demonstrated that multiple course sGCs can program altered hypothalamic-pituitary-adrenal (HPA) axis response to stress in first-generation (F1) and second-generation (F2) offspring. In this study, we sought to determine whether HPA axis activity and stress-associated behaviors (i.e., locomotor activity, attention) are altered after a single course of sGC in F1 and F2 female and male offspring. Pregnant guinea pigs [parental generation (F0)] received sGC (1 mg/kg) or saline on gestational days 50 and 51. HPA function and behavior were assessed in juvenile and adult F1 and F2 offspring of both sexes after maternal transmission. In F1, sGCs increased the HPA stress response in females but decreased responsiveness in males (P < 0.05). sGC exposure in F0 produced the opposite effects in F2 (P < 0.05). Reduced HPA responsiveness in F2 females was associated with reduced expression of proopiomelanocortin mRNA and increased expression of glucocorticoid receptor in the anterior pituitary (P < 0.05). Locomotor activity and prepulse inhibition were reduced by sGCs in adult F1 offspring. No behavioral changes were observed in F2 animals. These data indicate effects of antenatal treatment with a single course of sGC are present in F2 after maternal transmission. However, there are fewer effects on HPA activity and behavior in F1 and F2 offspring compared with treatment with multiple courses of sGCs.


Asunto(s)
Conducta Animal/efectos de los fármacos , Glucocorticoides/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Estrés Fisiológico/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Cobayas , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/fisiología , Patrón de Herencia/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología
6.
Epigenomics ; 10(4): 349-365, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29616589

RESUMEN

AIM: To determine the state of methylation of DNA molecules in the guinea pig hippocampus that are associated with either poised or active enhancers. METHODS: We used sequential chromatin immunoprecipitation-bisulfite-sequencing with an antibody to H3K4me1 to map the state of methylation of DNA that is found within enhancers. Actively transcribing transcription start sites were mapped by chromatin immunoprecipitation-sequencing with an antibody to RNApolII-PS5. Total DNA methylation was mapped using reduced representation bisulfite sequencing. RESULTS: DNA that overlaps with H3K4me1 binding regions in the genome is heavily methylated. However, DNA molecules that are found in H3K4me1 chromatin are hypomethylated, while DNA found in enhancers that are associated with active transcription is further demethylated. Differential methylation in enhancers is spotted in single CGs, bimodal and corresponds to transcription factor binding sites. CONCLUSION: Our study delineates the DNA methylation status of H3K4 me1-bound regions in the hippocampus in active and inactive genes.


Asunto(s)
Metilación de ADN , Elementos de Facilitación Genéticos , Hipocampo/metabolismo , Animales , Sitios de Unión , Genoma , Cobayas , Histonas/metabolismo , Masculino , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
7.
Sci Rep ; 7(1): 11814, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28924262

RESUMEN

Fetal exposure to high levels of glucocorticoids programs long-term changes in the physiologic stress response and behaviours. However, it is not known whether effects manifest in subsequent generations of offspring following maternal (MT) or paternal (PT) transmission. We treated pregnant guinea pigs with three courses of saline or synthetic glucocorticoid (sGC) at a clinically relevant dose. Altered cortisol response to stress and behaviours transmitted to juvenile female and male F2 and F3 offspring from both parental lines. Behavioural effects of sGC in F1-F3 PT females associated with altered expression of genes in the prefrontal cortex and hypothalamic paraventricular nucleus (PVN). Exposure to sGC programmed large transgenerational changes in PVN gene expression, including type II diabetes, thermoregulation, and collagen formation gene networks. We demonstrate transgenerational programming to F3 following antenatal sGC. Transmission is sex- and generation-dependent, occurring through both parental lines. Paternal transmission to F3 females strongly implicates epigenetic mechanisms of transmission.


Asunto(s)
Diabetes Mellitus Tipo 2 , Epigénesis Genética/efectos de los fármacos , Glucocorticoides/efectos adversos , Núcleo Hipotalámico Paraventricular , Efectos Tardíos de la Exposición Prenatal , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Glucocorticoides/farmacología , Cobayas , Masculino , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología
9.
J Steroid Biochem Mol Biol ; 160: 175-80, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26474822

RESUMEN

The embryo and fetus are highly responsive to the gestational environment. Glucocorticoids (GC) represent an important class of developmental cues and are crucial for normal brain development. Levels of GC in the fetal circulation are tightly regulated. They are maintained at low levels during pregnancy, and increase rapidly at the end of gestation. This surge in GC is critical for maturation of the organs, specifically the lungs, brain and kidney. There are extensive changes in brain epigenetic profiles that accompany the GC surge, suggesting that GC may drive regulation of gene transcription through altered epigenetic pathways. The epigenetic profiles produced by the GC surge can be prematurely induced as a result of maternal or fetal stress, as well as through exposure to synthetic glucocorticoids (sGC). This is highly clinically relevant as 10% of pregnant women are at risk for preterm labour and receive treatment with sGC to promote lung development in the fetus. Fetal overexposure to GC (including sGC) has been shown to cause lasting changes in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis leading to altered stress responses, and mood and anxiety disorders in humans and animals. In animal models, GC exposure is associated with transcriptomic and epigenomic changes that influence behaviour, HPA function and growth. Importantly, programming by GC results in sex-specific effects that can be inherited over multiple generations via paternal and maternal transmission.


Asunto(s)
Epigénesis Genética , Desarrollo Fetal , Glucocorticoides/metabolismo , Estrés Fisiológico , Animales , Femenino , Feto/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Embarazo
10.
Nat Rev Endocrinol ; 10(7): 391-402, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24863382

RESUMEN

Fetal development is a critical period for shaping the lifelong health of an individual. However, the fetus is susceptible to internal and external stimuli that can lead to adverse long-term health consequences. Glucocorticoids are an important developmental switch, driving changes in gene regulation that are necessary for normal growth and maturation. The fetal hypothalamic-pituitary-adrenal (HPA) axis is particularly susceptible to long-term programming by glucocorticoids; these effects can persist throughout the life of an organism. Dysfunction of the HPA axis as a result of fetal programming has been associated with impaired brain growth, altered behaviour and increased susceptibility to chronic disease (such as metabolic and cardiovascular disease). Moreover, the effects of glucocorticoid-mediated programming are evident in subsequent generations, and transmission of these changes can occur through both maternal and paternal lineages.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Glucocorticoides/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Adulto , Animales , Conducta Animal/efectos de los fármacos , Betametasona/efectos adversos , Niño , Dexametasona/efectos adversos , Femenino , Feto/efectos de los fármacos , Glucocorticoides/efectos adversos , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Embarazo , Nacimiento Prematuro/fisiopatología , Efectos Tardíos de la Exposición Prenatal , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología
11.
Nat Rev Endocrinol ; 10(7): 403-11, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24863383

RESUMEN

The lifelong health of an individual is shaped during critical periods of development. The fetus is particularly susceptible to internal and external stimuli, many of which can alter developmental trajectories and subsequent susceptibility to disease. Glucocorticoids are critical in normal development of the fetus, as they are involved in the growth and maturation of many organ systems. The surge in fetal glucocorticoid levels that occurs in most mammalian species over the last few days of pregnancy is an important developmental switch leading to fundamental changes in gene regulation in many organs, including the brain. These changes are important for the transition to postnatal life. Exposure of the fetus to increased levels of glucocorticoids, resulting from maternal stress or treatment with synthetic glucocorticoids, can lead to long-term 'programming' of hypothalamic-pituitary-adrenal function and behaviours. Glucocorticoids act at multiple levels within the fetal brain. Growing evidence indicates that they can exert powerful effects on the epigenome, including on DNA methylation, histone acetylation and microRNA, to influence gene expression. Such influences probably represent a critical component of the 'programming' process, and might be partly responsible for the transgenerational effects of antenatal glucocorticoid exposure on neurologic, cardiovascular and metabolic function.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Glucocorticoides/fisiología , Sistema Hipotálamo-Hipofisario/embriología , Sistema Hipófiso-Suprarrenal/embriología , Animales , Encéfalo/embriología , Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Intercambio Materno-Fetal , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/complicaciones
12.
Endocrinology ; 154(11): 4170-81, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029241

RESUMEN

The endogenous glucocorticoid (GC) surge in late gestation plays a vital role in maturation of several organ systems. For this reason, pregnant women at risk of preterm labor are administered synthetic glucocorticoids (sGCs) to promote fetal lung development. Animal studies have shown that fetal sGC exposure can cause life-long changes in endocrine and metabolic function. We have previously shown that antenatal sGC treatment is associated with alterations in global DNA methylation and modifications to the hippocampal methylome and acetylome. In this study, we hypothesized that: 1) there are changes in the transcriptional landscape of the fetal hippocampus in late gestation, associated with the endogenous cortisol surge; 2) fetal sGC exposure alters genome-wide transcription in the hippocampus; and 3) these changes in transcription are associated with modified glucocorticoid receptor (GR) DNA binding and DNA methylation. sGC was administered as 2 courses on gestational days (GD) 40, 41, 50, and 51, and the hippocampi of fetal guinea pigs were examined before (GD52) and after (GD65) the endogenous cortisol surge (Term ∼GD67). We also analyzed fetal hippocampi 24 hours and 14 days following maternal sGC injections (n = 3-4/group). Genome-wide modification of transcription and GR DNA binding occurred in late gestation, in parallel with the normal GC surge. Further, sGC exposure had a substantial impact on the hippocampal transcriptome, GR-DNA binding, and DNA methylation at 24 hours and 14 days following the final sGC treatment. These data support the hypothesis that GC exposure in late gestation plays a significant role in modifying the transcriptional and epigenetic landscape of the developing fetal hippocampus and that substantial effects are evident for at least 2 weeks after sGC exposure.


Asunto(s)
Betametasona/farmacología , Metilación de ADN , Hipocampo/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Betametasona/administración & dosificación , Femenino , Desarrollo Fetal/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Genoma , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Cobayas , Hipocampo/embriología , Masculino , Embarazo , ARN Mensajero/genética , Receptores de Glucocorticoides/genética , Transcriptoma
13.
Endocrinology ; 154(3): 1168-80, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23389956

RESUMEN

The late-gestation surge in fetal plasma cortisol is critical for maturation of fetal organ systems. As a result, synthetic glucocorticoids (sGCs) are administered to pregnant women at risk of delivering preterm. However, animal studies have shown that fetal exposure to sGC results in increased risk of behavioral, endocrine, and metabolic abnormalities in offspring. Here, we test the hypothesis that prenatal GC exposure resulting from the fetal cortisol surge or after sGC exposure results in promoter-specific epigenetic changes in the hippocampus. Fetal guinea pig hippocampi were collected before (gestational day [GD52]) and after (GD65) the fetal plasma cortisol surge (Term∼GD67) and 24 hours after (GD52) and 14 days after (GD65) two repeat courses of maternal sGC (betamethasone) treatment (n = 3-4/gp). We identified extensive genome-wide alterations in promoter methylation in late fetal development (coincident with the fetal cortisol surge), whereby the majority of the affected promoters exhibited hypomethylation. Fetuses exposed to sGC in late gestation exhibited substantial differences in DNA methylation and histone h3 lysine 9 (H3K9) acetylation in specific gene promoters; 24 hours after the sGC treatment, the majority of genes affected were hypomethylated or hyperacetylated. However, 14 days after sGC exposure these differences did not persist, whereas other promoters became hypermethylated or hyperacetylated. These data support the hypothesis that the fetal GC surge is responsible, in part, for significant variations in genome-wide promoter methylation and that prenatal sGC treatment profoundly changes the epigenetic landscape, affecting both DNA methylation and H3K9 acetylation. This is important given the widespread use of sGC in the management of women in preterm labor.


Asunto(s)
Epigénesis Genética , Feto/metabolismo , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Animales , Metilación de ADN , Femenino , Desarrollo Fetal/genética , Cobayas , Hipocampo/embriología , Histonas/metabolismo , Hidrocortisona/sangre , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
PLoS One ; 7(8): e43022, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22973436

RESUMEN

Placental P-glycoprotein (P-gp) acts to protect the developing fetus from exogenous compounds. This protection declines with advancing gestation leaving the fetus and fetal brain vulnerable to these compounds and potential teratogens in maternal circulation. This vulnerability may be more pronounced in pregnancies complicated by infection, which is common during pregnancy. Pro-inflammatory cytokines (released during infection) have been shown to be potent inhibitors of P-gp, but nothing is known regarding their effects at the developing blood-brain barrier (BBB). We hypothesized that P-gp function and expression in endothelial cells of the developing BBB will be inhibited by pro-inflammatory cytokines. We have derived brain endothelial cell (BEC) cultures from various stages of development of the guinea pig: gestational day (GD) 50, 65 (term ~68 days) and postnatal day (PND) 14. Once these cultures reached confluence, BECs were treated with various doses (10(0)-10(4 )pg/mL) of pro-inflammatory cytokines: interleukin-1ß (IL-1ß), interleukin-6 (IL-6) or tumor necrosis factor- α (TNF-α). P-gp function or abcb1 mRNA (encodes P-gp) expression was assessed following treatment. Incubation of GD50 BECs with IL-1ß, IL-6 or TNF-α resulted in no change in P-gp function. GD65 BECs displayed a dose-dependent decrease in function with all cytokines tested; maximal effects at 42%, 65% and 34% with IL-1ß, IL-6 and TNF-α treatment, respectively (P<0.01). Inhibition of P-gp function by IL-1ß, IL-6 and TNF-α was even greater in PND14 BECs; maximal effects at 36% (P<0.01), 84% (P<0.05) and 55% (P<0.01), respectively. Cytokine-induced reductions in P-gp function were associated with decreased abcb1 mRNA expression. These data suggest that BBB P-gp function is increasingly responsive to the inhibitory effects of pro-inflammatory cytokines, with increasing developmental age. Thus, women who experience infection and take prescription medication during pregnancy may expose the developing fetal brain to greater amounts of exogenous compounds - many of which are considered potentially teratogenic.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Citocinas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cobayas , Interleucina-1beta/farmacología , Interleucina-6/farmacología , Masculino , Embarazo , Factor de Necrosis Tumoral alfa/farmacología
15.
Endocrinology ; 153(7): 3295-307, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22564976

RESUMEN

Approximately 10% of pregnant women are at risk of preterm delivery and receive synthetic glucocorticoids (sGC) to promote fetal lung development. Studies have indicated that prenatal sGC therapy modifies hypothalamic-pituitary-adrenal (HPA) function in first-generation (F(1)) offspring. The objective of this study was to determine whether differences in HPA function and behavior are evident in the subsequent (F(2)) generation. Pregnant guinea pigs (F(0)) received betamethasone (BETA; 1 mg/kg) or saline on gestational d 40/41, 50/51, and 60/61. F(1) females were mated with control males to create F(2) offspring. HPA function was assessed in juvenile and adult F(2) offspring. Locomotor activity was assessed in juvenile offspring. Analysis of HPA-related gene expression was undertaken in adult hippocampi, hypothalami, and pituitaries. Locomotor activity was reduced in F(2) BETA males (P < 0.05). F(2) BETA offspring displayed blunted cortisol response to swim stress (P < 0.05). After dexamethasone challenge, F(2) BETA males and females displayed increased and decreased negative feedback, respectively. F(2) BETA females had reduced pituitary levels of proopiomelanocortin (and adrenocorticotropic hormone), and corticotropin-releasing hormone receptor mRNA and protein (P < 0.05). F(2) BETA males displayed increased hippocampal glucocorticoid receptor (P < 0.001), whereas in BETA females, hippocampal glucocorticoid receptor and mineralocorticoid receptor mRNA were decreased (P < 0.05). In conclusion, prenatal BETA treatment affects HPA function and behavior in F(2) offspring. In F(2) BETA females, pituitary function appears to be primarily affected, whereas hippocampal glucocorticoid feedback systems appear altered in both F(2) BETA males and females. These data have clinical implication given the widespread use of repeat course glucocorticoid therapy in the management of preterm labour.


Asunto(s)
Glucocorticoides/uso terapéutico , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Animales , Betametasona/farmacología , Dexametasona/farmacología , Epigénesis Genética , Femenino , Cobayas , Hidrocortisona/metabolismo , Hibridación in Situ , Masculino , Exposición Materna , Modelos Biológicos , Embarazo , Preñez , Efectos Tardíos de la Exposición Prenatal , Saliva/metabolismo
16.
Endocrinology ; 153(7): 3269-83, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22564977

RESUMEN

Prenatal synthetic glucocorticoids (sGC) are administered to pregnant women at risk of delivering preterm, approximately 10% of all pregnancies. Animal studies have demonstrated that offspring exposed to elevated glucocorticoids, either by administration of sGC or as a result of maternal stress, are at increased risk of developing behavioral, endocrine, and metabolic abnormalities. DNA methylation is a covalent modification of DNA that plays a critical role in long-lasting programming of gene expression. Here we tested the hypothesis that prenatal sGC treatment has both acute and long-term effects on DNA methylation states in the fetus and offspring and that these effects extend into a subsequent generation. Pregnant guinea pigs were treated with sGC in late gestation, and methylation analysis by luminometric methylation assay was undertaken in organs from fetuses and offspring across two generations. Expression of genes that modify the epigenetic state were measured by quantitative real-time PCR. Results indicate that there are organ-specific developmental trajectories of methylation in the fetus and newborn. Furthermore, these trajectories are substantially modified by intrauterine exposure to sGC. These sGC-induced changes in DNA methylation remain into adulthood and are evident in the next generation. Furthermore, prenatal sGC exposure alters the expression of several genes encoding proteins that modulate the epigenetic state. Several of these changes are long lasting and are also present in the next generation. These data support the hypothesis that prenatal sGC exposure leads to broad changes in critical components of the epigenetic machinery and that these effects can pass to the next generation.


Asunto(s)
Metilación de ADN , Glucocorticoides/metabolismo , Animales , Cruzamientos Genéticos , ADN/metabolismo , Epigénesis Genética , Femenino , Cobayas , Masculino , Exposición Materna , Fenotipo , Embarazo , Preñez , Efectos Tardíos de la Exposición Prenatal , ARN Mensajero/metabolismo , Riesgo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA