Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Aging ; 61: 13-22, 2018 01.
Article in English | MEDLINE | ID: mdl-29031089

ABSTRACT

Alternative RNA splicing results in the translation of diverse protein products arising from a common nucleotide sequence. These alternative protein products are often functional and can have widely divergent actions from the canonical protein. Studies in humans and other vertebrate animals have demonstrated that alternative splicing events increase with advanced age, sometimes resulting in pathological consequences. Menopause represents a critical transition for women, where the beneficial effects of estrogens are no longer evident; therefore, factors underlying increased pathological conditions in women are confounded by the dual factors of aging and declining estrogens. Estrogen receptors (ERs) are subject to alternative splicing, the spliced variants increase following menopause, and they fail to efficiently activate estrogen-dependent signaling pathways. However, the factors that regulate the alternative splicing of ERs remain unknown. We demonstrate novel evidence supporting a potential biological feedback loop where 17ß-estradiol regulates the RNA-binding protein Nova1, which, in turn, regulates the alternative splicing of ERß. These data increase our understanding of ER alternative splicing and could have potential implications for women taking hormone replacement therapy after menopause.


Subject(s)
Aging/genetics , Aging/metabolism , Alternative Splicing/genetics , Brain/metabolism , Estradiol/physiology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Gene Expression Regulation , RNA-Binding Proteins/physiology , Animals , Cells, Cultured , Female , Humans , Neuro-Oncological Ventral Antigen , Rats, Inbred F344
2.
Alcohol ; 60: 179-189, 2017 05.
Article in English | MEDLINE | ID: mdl-27817987

ABSTRACT

Teenage binge drinking is a major health concern in the United States, with 21% of teenagers reporting binge-pattern drinking behavior in the previous 30 days. Recently, our lab showed that alcohol-naïve offspring of rats exposed to alcohol during adolescence exhibited altered gene expression profiles in the hypothalamus, a brain region involved in stress regulation. We employed Enhanced Reduced Representation Bisulfite Sequencing as an unbiased approach to test the hypothesis that parental exposure to binge-pattern alcohol during adolescence alters DNA methylation profiles in their alcohol-naïve offspring. Wistar rats were administered a repeated binge-ethanol exposure paradigm during early (postnatal day (PND) 37-44) and late (PND 67-74) adolescent development. Animals were mated 24 h after the last ethanol dose and subsequent offspring were produced. Analysis of male PND7 offspring revealed that offspring of alcohol-exposed parents exhibited differential DNA methylation patterns in the hypothalamus. The differentially methylated cytosines (DMCs) were distinct between offspring depending on which parent was exposed to ethanol. Moreover, novel DMCs were observed when both parents were exposed to ethanol and many DMCs from single parent ethanol exposure were not recapitulated with dual parent exposure. We also measured mRNA expression of several differentially methylated genes and some, but not all, showed correlative changes in expression. Importantly, methylation was not a direct predictor of expression levels, underscoring the complexity of transcriptional regulation. Overall, we demonstrate that adolescent binge ethanol exposure causes altered genome-wide DNA methylation patterns in the hypothalamus of alcohol-naïve offspring.


Subject(s)
Binge Drinking/genetics , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Ethanol/toxicity , Hypothalamus/drug effects , Inheritance Patterns , Underage Drinking , Age Factors , Animals , Binge Drinking/metabolism , Female , Gene Expression Regulation , Heredity , Hypothalamus/metabolism , Male , Models, Animal , Pedigree , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Sexual Development , Time Factors
3.
PLoS One ; 11(8): e0160276, 2016.
Article in English | MEDLINE | ID: mdl-27487271

ABSTRACT

Aging and the coincident loss of circulating estrogens at menopause lead to increased risks for neurological and cardiovascular pathologies. Clinical studies show that estrogen therapy (ET) can be beneficial in mitigating these negative effects, in both the brain and heart, when it is initiated shortly after the perimenopausal transition. However, this same therapy is detrimental when initiated >10 years postmenopause. Importantly, the molecular mechanisms underlying this age-related switch in ET efficacy are unknown. Estrogen receptors (ERs) mediate the neuroprotective and cardioprotective functions of estrogens by modulating gene transcription or, non-genomically, by activating second messenger signaling pathways, such as mitogen activated protein kinases (MAPK). These kinases are critical regulators of cell signaling pathways and have widespread downstream effects. Our hypothesis is that age and estrogen deprivation following menopause alters the expression and activation of the MAPK family members p38 and ERK in the brain and heart. To test this hypothesis, we used a surgically induced model of menopause in 18 month old rats through bilateral ovariectomy (OVX) followed by an acute dose of 17ß-estradiol (E2) administered at varying time points post-OVX (1 week, 4 weeks, 8 weeks, or 12 weeks). Age and E2 treatment differentially regulated kinase activity in both the brain and heart, and the effects were also brain region specific. MAPK signaling plays an integral role in aging, and the aberrant regulation of those signaling pathways might be involved in age-related disorders. Clinical studies show benefits of ET during early menopause but detrimental effects later, which might be reflective of changes in kinase expression and activation status.


Subject(s)
Aging/physiology , Brain/drug effects , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/drug effects , MAP Kinase Signaling System/drug effects , Aging/drug effects , Animals , Brain/metabolism , Female , Myocardium/metabolism , Ovariectomy , Rats , Rats, Inbred F344 , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Oncotarget ; 6(35): 36965-83, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26460619

ABSTRACT

Administration of 17ß-estradiol (E2) has beneficial effects on cognitive function in peri- but not post-menopausal women, yet the molecular mechanisms underlying age-related changes in E2 action remain unclear. We propose that there is a biological switch in E2 action that occurs coincident with age and length of time after ovarian hormone depletion, and we hypothesized that age-dependent regulation of microRNAs (miRNAs) could be the molecular basis for that switch. Previously we showed that miRNAs are regulated by E2 in young compared to aged female rats. Here we tested whether increasing lengths of ovarian hormone deprivation in aged females altered E2 regulation of these mature miRNAs. In addition, we determined where along the miRNA biogenesis pathway E2 exerted its effects. Our results showed that age and increased lengths of ovarian hormone deprivation abolished the ability of E2 to regulate mature miRNA expression in the brain. Further, we show that E2 acted at specific points along the miRNA biogenesis pathway.


Subject(s)
Aging , Brain/metabolism , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hypothalamus/metabolism , MicroRNAs/genetics , Animals , Blotting, Western , Brain/drug effects , Brain/pathology , Female , Gene Expression Profiling , Hypothalamus/drug effects , Hypothalamus/pathology , Ovariectomy , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
Endocrinology ; 156(11): 4187-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26295370

ABSTRACT

Loss of circulating 17ß-estradiol (E2) that occurs during menopause can have detrimental effects on cognitive function. The efficacy of hormone replacement therapy declines as women become farther removed from the menopausal transition, yet the molecular mechanisms underlying this age-related switch in E2 efficacy are unknown. We hypothesized that aging and varying lengths of E2 deprivation alters the ratio of alternatively spliced estrogen receptor (ER)ß isoforms in the brain of female rats. Further, we tested whether changes in global transcriptional activity and splicing kinetics regulate the alternative splicing of ERß. Our results revealed brain region-specific changes in ERß alternative splicing in both aging and E2-deprivation paradigms and showed that ERß could mediate E2-induced alternative splicing. Global transcriptional activity, as measured by phosphorylated RNA polymerase II, was also regulated by age and E2 in specific brain regions. Finally, we show that inhibition of topoisomerase I resulted in increased ERß2 splice variant expression.


Subject(s)
Aging/metabolism , Alternative Splicing , Brain/metabolism , Estradiol/blood , Estrogen Receptor beta/metabolism , Protein Isoforms/metabolism , Aging/genetics , Animals , Estrogen Receptor beta/genetics , Female , Phosphorylation , Protein Isoforms/genetics , Rats , Rats, Inbred F344 , Transcription, Genetic , Transcriptional Activation
6.
Mol Cell Proteomics ; 13(3): 760-79, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390426

ABSTRACT

Recent clinical evidence suggests that the neuroprotective and beneficial effects of hormone therapy may be limited by factors related to age and reproductive status. The patient's age and length of time without circulating ovarian hormones are likely to be key factors in the specific neurological outcomes of hormone therapy. However, the mechanisms underlying age-related changes in hormone efficacy have not been determined. We hypothesized that there are intrinsic changes in estrogen receptor ß (ERß) function that determine its ability to mediate the actions of 17ß-estradiol (E2) in brain regions such as the ventral hippocampus. In this study, we identified and quantified a subset of ERß protein interactions in the ventral hippocampus that were significantly altered by E2 replacement in young and aged animals, using two-dimensional differential gel electrophoresis coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry. This study demonstrates quantitative changes in ERß protein-protein interactions with E2 replacement that are dependent upon age in the ventral hippocampus and how these changes could alter processes such as transcriptional regulation. Thus, our data provide evidence that changes in ERß protein interactions are a potential mechanism for age-related changes in E2 responsiveness in the brain after menopause.


Subject(s)
Aging/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Hippocampus/metabolism , Protein Interaction Mapping , Adenosine Triphosphatases/metabolism , Aging/drug effects , Animals , Annexin A5/metabolism , Cell Cycle Proteins/metabolism , Estrogen Receptor beta/genetics , Female , Gelsolin/metabolism , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HEK293 Cells , Hippocampus/drug effects , Humans , Image Processing, Computer-Assisted , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Response Elements/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...