Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 21082, 2023 11 30.
Article in English | MEDLINE | ID: mdl-38030664

ABSTRACT

The genetic material encoded on X and Y chromosomes provides the foundation by which biological sex differences are established. Epigenetic regulators expressed on these sex chromosomes, including Kdm6a (Utx), Kdm5c, and Ddx3x have far-reaching impacts on transcriptional control of phenotypic sex differences. Although the functionality of UTY (Kdm6c, the Y-linked homologue of UTX), has been supported by more recent studies, its role in developmental sex differences is not understood. Here we test the hypothesis that UTY is an important transcriptional regulator during development that could contribute to sex-specific phenotypes and disease risks across the lifespan. We generated a random insertion Uty transgenic mouse (Uty-Tg) to overexpress Uty. By comparing transcriptomic profiles in developmental tissues, placenta and hypothalamus, we assessed potential UTY functional activity, comparing Uty-expressing female mice (XX + Uty) with wild-type male (XY) and female (XX) mice. To determine if Uty expression altered physiological or behavioral outcomes, adult mice were phenotypically examined. Uty expression masculinized female gene expression patterns in both the placenta and hypothalamus. Gene ontology (GO) and gene set enrichment analysis (GSEA) consistently identified pathways including immune and synaptic signaling as biological processes associated with UTY. Interestingly, adult females expressing Uty gained less weight and had a greater glucose tolerance compared to wild-type male and female mice when provided a high-fat diet. Utilizing a Uty-overexpressing transgenic mouse, our results provide novel evidence as to a functional transcriptional role for UTY in developing tissues, and a foundation to build on its prospective capacity to influence sex-specific developmental and health outcomes.


Subject(s)
Gene Expression Regulation , Transcriptome , Male , Female , Animals , Mice , Prospective Studies , Gene Expression Profiling , Mice, Transgenic
2.
AIDS ; 37(6): 895-903, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36728423

ABSTRACT

OBJECTIVES: To examine female participation and the observed efficacy and safety by sex from phase 3 HIV-1 trials submitted to the United States Food and Drug Administration (FDA) to support approval or a major labeling change. DESIGN: Our analyses were based on phase 3 trials in HIV-1 infected treatment-naive adults submitted to FDA since 2010. METHODS: We evaluated enrollment of treatment-naive females in 18 clinical trials for HIV-1. Participation to prevalence ratio (PPR) was calculated as the percentage of females among trial participants divided by the percentage of females in the disease population. PPR between 0.8 and 1.2 reflects similar representation of females in the trial and the disease population. Sex differences in efficacy (virologic response rates) and selected safety events were evaluated. RESULTS: United States (US) females, particularly US Black females were not adequately represented in clinical trials. The PPR for US females overall was 0.59 and for US Black females was 0.63. Statistically significant sex differences favoring males were observed for efficacy outcomes in both the global population and US participants. Statistically significant sex differences were observed for some safety outcomes. CONCLUSIONS: US females are underrepresented in phase 3 HIV-1 clinical trials. Underrepresentation was not likely due to enrollment criteria. Statistically significant sex differences were noted for efficacy and selected safety outcomes; however, some differences were not clinically relevant. The ability to detect sex differences was hindered by low numbers of female participants overall and within subgroups. Additional research into innovative approaches to recruit and retain females in clinical trials should continue.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Adult , Humans , Male , Female , United States , Sex Characteristics , HIV Infections/drug therapy , HIV Seropositivity/drug therapy , Pharmaceutical Preparations
3.
Cells ; 10(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34685591

ABSTRACT

Sexual selection arising from sperm competition has driven the evolution of immense variation in ejaculate allocation and sperm characteristics not only among species, but also among males within a species. One question that has received little attention is how cooperation among males affects these patterns. Here we ask how male alternative reproductive types differ in testes size, ejaculate production, and sperm morphology in the ocellated wrasse, a marine fish in which unrelated males cooperate and compete during reproduction. Nesting males build nests, court females and provide care. Sneaker males only "sneak" spawn, while satellite males sneak, but also help by chasing away sneakers. We found that satellite males have larger absolute testes than either sneakers or nesting males, despite their cooperative role. Nesting males invested relatively less in testes than either sneakers or satellites. Though sneakers produced smaller ejaculates than either satellite or nesting males, we found no difference among male types in either sperm cell concentration or sperm number, implying sneakers may produce less seminal fluid. Sperm tail length did not differ significantly among male types, but sneaker sperm cells had significantly larger heads than either satellite or nesting male sperm, consistent with past research showing sneakers produce slower sperm. Our results highlight that social interactions among males can influence sperm and ejaculate production.


Subject(s)
Fishes/metabolism , Reproduction , Spermatozoa/metabolism , Testis/metabolism , Animals , Male
4.
Pharm Stat ; 20(5): 929-938, 2021 09.
Article in English | MEDLINE | ID: mdl-34396690

ABSTRACT

Differences in patient characteristics, including age, sex, and race influence the safety and effectiveness of drugs, biologic products, and medical devices. Here we provide a summary of the topics discussed during the opening panel at the 2018 Johns Hopkins Center for Excellence in Regulatory Science and Innovation symposium on Assessing and Communicating Heterogeneity of Treatment Effects for Patient Subpopulations: Challenges and Opportunities. The goal of this session was to provide a brief overview of FDA-regulated therapeutics, including drugs, biologics and medical devices, and some of the major sources of heterogeneity of treatment effects (HTE) related to patient demographics, such as age, sex and race. The panel discussed the US Food and Drug Administration's role in reviewing and regulating drugs, devices, and biologic products and the challenges associated with ensuring that diverse patient populations benefit from these therapeutics. Ultimately, ensuring diverse demographic inclusion in clinical trials, and designing basic and clinical research studies to account for the intended patient population's age, sex, race, and genetic factors among other characteristics, will lead to better, safer therapies for diverse patient populations.


Subject(s)
Pharmaceutical Preparations , United States Food and Drug Administration , Humans , United States
5.
Med ; 2(5): 456-459, 2021 05 14.
Article in English | MEDLINE | ID: mdl-35590224

ABSTRACT

Recruiting and retaining diverse populations in clinical research is critically important. Over the years, we have seen improvements in the representation of women in clinical trials submitted in FDA marketing applications, and we are encouraged by the potential for new strategies to further their representation.


Subject(s)
Marketing , Female , Humans
6.
Placenta ; 100: 164-170, 2020 10.
Article in English | MEDLINE | ID: mdl-32980048

ABSTRACT

INTRODUCTION: Despite a wealth of epidemiological evidence that cumulative parental lifetime stress experiences prior to conception are determinant of offspring developmental trajectories, there is a lack of insight on how these previous stress experiences are stored and communicated intergenerationally. Preconception experiences may impact offspring development through alterations in transcriptional regulation of the placenta, a major determinant of offspring growth and sex-specific developmental outcomes. We evaluated the lasting influence of maternal and paternal preconception stress (PCS) on the mid-gestation placenta and fetal brain, utilizing their transcriptomes as proximate readouts of intergenerational impact. METHODS: To assess the combined vs. dominant influence of maternal and paternal preconception environment on sex-specific fetal development, we compared transcriptional outcomes using a breeding scheme of one stressed parent, both stressed parents, or no stressed parents as controls. RESULTS: Interestingly, offspring sex affected the directionality of transcriptional changes in response to PCS, where male tissues showed a predominant downregulation, and female tissues showed an upregulation. There was also an intriguing effect of parental sex on placental programming where paternal PCS drove more effects in female placentas, while maternal PCS produced more transcriptional changes in male placentas. However, in the fetal brain, maternal PCS produced overall more changes in gene expression than paternal PCS, supporting the idea that the intrauterine environment may have a larger overall influence on the developing brain than it does on shaping the placenta. DISCUSSION: Preconception experiences drive changes in the placental and the fetal brain transcriptome at a critical developmental timepoint. While not determinant, these altered transcriptional states may underlie sex-biased risk or resilience to stressful experiences later in life.


Subject(s)
Brain/metabolism , Fetus/metabolism , Placenta/metabolism , Preconception Injuries , Stress, Psychological , Animals , Female , Male , Mice , Pregnancy , Sex Characteristics , Transcriptome
7.
Nat Commun ; 11(1): 1499, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198406

ABSTRACT

Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of incredible specificity in transmitting signals involved in cellular function, including germ cell maturation. Spermatogenesis occurs in the testes, behind a protective barrier to ensure safeguarding of germline DNA from environmental insults. Following DNA compaction, further sperm maturation occurs in the epididymis. Here, we report reproductive tract EVs transmit information regarding stress in the paternal environment to sperm, potentially altering fetal development. Using intracytoplasmic sperm injection, we found that sperm incubated with EVs collected from stress-treated epididymal epithelial cells produced offspring with altered neurodevelopment and adult stress reactivity. Proteomic and transcriptomic assessment of these EVs showed dramatic changes in protein and miRNA content long after stress treatment had ended, supporting a lasting programmatic change in response to chronic stress. Thus, EVs as a normal process in sperm maturation, can also perform roles in intergenerational transmission of paternal environmental experience.


Subject(s)
Extracellular Vesicles/metabolism , Nervous System/growth & development , Proteomics , Reproduction/physiology , Adolescent , Animals , Cell Culture Techniques , Epididymis/metabolism , Epigenesis, Genetic , Epigenomics , Female , Germ Cells , Histones , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Nanoparticles , Sperm Maturation/genetics , Sperm Maturation/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatozoa/metabolism , Stress, Physiological , Testis
8.
Mol Ecol ; 28(15): 3464-3481, 2019 08.
Article in English | MEDLINE | ID: mdl-30586201

ABSTRACT

While extensive research has focused on how social interactions evolve, the fitness consequences of the neuroendocrine mechanisms underlying these interactions have rarely been documented, especially in the wild. Here, we measure how the neuroendocrine mechanisms underlying male behaviour affect mating success and sperm competition in the ocellated wrasse (Symphodus ocellatus). In this species, males exhibit three alternative reproductive types. "Nesting males" provide parental care, defend territories and form cooperative associations with unrelated "satellites," who cheat by sneaking fertilizations but help by reducing sperm competition from "sneakers" who do not cooperate or provide care. To measure the fitness consequences of the mechanisms underlying these social interactions, we used "phenotypic engineering" that involved administering an androgen receptor antagonist (flutamide) to wild, free-living fish. Nesting males treated with flutamide shifted their aggression from sneakers to satellite males and experienced decreased submissiveness by sneaker males (which correlated with decreased nesting male mating success). The preoptic area (POA), a region controlling male reproductive behaviours, exhibited dramatic down-regulation of androgen receptor (AR) and vasotocin 1a receptor (V1aR) mRNA following experimental manipulation of androgen signalling. We did not find a direct effect of the manipulation on male mating success, paternity or larval production. However, variation in neuroendocrine mechanisms generated by the experimental manipulation was significantly correlated with changes in behaviour and mating success: V1aR expression was negatively correlated with satellite-directed aggression, and expression of its ligand arginine vasotocin (AVT) was positively correlated with courtship and mating success, thus revealing the potential for sexual selection on these mechanisms.


Subject(s)
Neurosecretory Systems/physiology , Perciformes/physiology , Sexual Behavior, Animal/physiology , Social Behavior , Spermatozoa/physiology , Androgen Receptor Antagonists/pharmacology , Animals , Brain/metabolism , Female , Flutamide/pharmacology , Male , Neurosecretory Systems/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Reproduction/drug effects , Reproduction/physiology , Sexual Behavior, Animal/drug effects , Spermatozoa/drug effects , Testosterone/analogs & derivatives , Testosterone/metabolism
9.
Nat Commun ; 9(1): 2555, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29967448

ABSTRACT

Although sex biases in disease presentation are well documented, the mechanisms mediating vulnerability or resilience to diseases are unknown. In utero insults are more likely to produce detrimental health outcomes for males versus females. In our mouse model of prenatal stress, male offspring experience long-term dysregulation of body weight and hypothalamic pituitary adrenal stress axis dysfunction, endophenotypes of male-biased neurodevelopmental disorders. Placental function is critical for healthy fetal development, and we previously showed that sex differences in placental O-linked N-acetylglucosamine transferase (OGT) mediate the effects of prenatal stress on neurodevelopmental programming. Here we show that one mechanism whereby sex differences in OGT confer variation in vulnerability to prenatal insults is by establishing sex-specific trophoblast gene expression patterns and via regulation of the canonically repressive epigenetic modification, H3K27me3. We demonstrate that high levels of H3K27me3 in the female placenta create resilience to the altered hypothalamic programming associated with prenatal stress exposure.


Subject(s)
Histones/metabolism , N-Acetylglucosaminyltransferases/physiology , Placenta/metabolism , Prenatal Exposure Delayed Effects/pathology , Stress, Physiological/physiology , Animals , Body Weight , Disease Models, Animal , Embryo, Mammalian , Epigenesis, Genetic/physiology , Female , Fetal Development/physiology , Gene Expression Profiling , Genes, X-Linked/physiology , Histone Code/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Restraint, Physical , Sex Factors , Trophoblasts/metabolism
10.
Biol Psychiatry ; 83(10): 886-894, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29198470

ABSTRACT

Parental stress exposures are implicated in the risk for offspring neurodevelopmental and neuropsychiatric disorders, prompting critical examination of preconception and prenatal periods as vulnerable to environmental insults such as stress. Evidence from human studies and animal models demonstrates the influence that both maternal and paternal stress exposures have in changing the course of offspring brain development. Mechanistic examination of modes of intergenerational transmission of exposure during pregnancy has pointed to alterations in placental signaling, including changes in inflammatory, nutrient-sensing, and epigenetic pathways. Transmission of preconception paternal stress exposure is associated with changes in epigenetic marks in sperm, with a primary focus on the reprogramming of DNA methylation, histone posttranslational modifications, and small noncoding RNAs. In this review, we discuss evidence supporting the important contribution of intergenerational parental stress in offspring neurodevelopment and disease risk, and the currently known epigenetic mechanisms underlying this transmission.


Subject(s)
Neurodevelopmental Disorders/etiology , Parent-Child Relations , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/complications , Female , Humans , Parents , Pregnancy
11.
Nat Rev Neurosci ; 18(8): 471-484, 2017 08.
Article in English | MEDLINE | ID: mdl-28638119

ABSTRACT

The study of sex differences in the brain is a topic of neuroscientific study that has broad reaching implications for culture, society and biomedical science. Recent research in rodent models has led to dramatic shifts in our views of the mechanisms underlying the sexual differentiation of the brain. These include the surprising discoveries of a role for immune cells and inflammatory mediators in brain masculinization and a role for epigenetic suppression in brain feminization. How and to what degree these findings will translate to human brain development will be questions of central importance in future research in this field.


Subject(s)
Brain/growth & development , Brain/metabolism , Epigenesis, Genetic/physiology , Immune System/physiology , Inflammation Mediators/physiology , Sex Characteristics , Sex Differentiation/physiology , Animals , Humans
13.
Brain Behav Evol ; 89(3): 153-161, 2017.
Article in English | MEDLINE | ID: mdl-28448987

ABSTRACT

Oxytocin (OT) mediates social habituation in rodent model systems, but its role in mediating this effect in other vertebrates is unknown. We used males of the African cichlid fish, Astatotilapia burtoni, to investigate two aspects of isotocin (IT; an OT homolog) signaling in social habituation. First, we examined the expression of IT receptor 2 (ITR2) as well as two immediate early genes in brain regions implicated in social recognition. Next, we examined IT neuron activity using immunohistochemistry. Patterns of gene expression in homologs of the amygdala and hippocampus implicate IT signaling in these regions in social habituation to a territorial neighbor. In the preoptic area, the expression of the ITR2 subtype and IT neuron activity respond to the presence of a male, independent of familiarity. Our results implicate IT in mediating social habituation in a teleost.


Subject(s)
Cichlids/genetics , Oxytocin/analogs & derivatives , Aggression/physiology , Amygdala , Animals , Behavior, Animal/physiology , Brain/metabolism , Cichlids/physiology , Female , In Situ Hybridization , Male , Oxytocin/metabolism , Oxytocin/physiology , Preoptic Area/metabolism , Receptors, Oxytocin/physiology , Social Behavior
14.
Mol Ecol ; 26(2): 505-518, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27862511

ABSTRACT

Gene expression differences between males and females often underlie sexually dimorphic phenotypes, and the expression levels of genes that are differentially expressed between the sexes are thought to respond to sexual selection. Most studies on the transcriptomic response to sexual selection treat sexual selection as a single force, but postmating sexual selection in particular is expected to specifically target gonadal tissue. The three male morphs of the ocellated wrasse (Symphodus ocellatus) make it possible to test the role of postmating sexual selection in shaping the gonadal transcriptome. Nesting males hold territories and have the highest reproductive success, yet we detected feminization of their gonadal gene expression compared to satellite males. Satellite males are less brightly coloured and experience more intense sperm competition than nesting males. In line with postmating sexual selection affecting gonadal gene expression, we detected a more masculinized expression profile in satellites. Sneakers are the lowest quality males and showed both de-masculinization and de-feminization of gene expression. We also detected higher rates of gene sequence evolution of male-biased genes compared to unbiased genes, which could at least in part be explained by positive selection. Together, these results reveal the potential for postmating sexual selection to drive higher rates of gene sequence evolution and shape the gonadal transcriptome profile.


Subject(s)
Perciformes/genetics , Reproduction , Spermatozoa/physiology , Transcriptome , Animals , Evolution, Molecular , Female , Male , Phenotype , Sexual Behavior, Animal
15.
Front Behav Neurosci ; 9: 221, 2015.
Article in English | MEDLINE | ID: mdl-26347630

ABSTRACT

The notion that epigenetics may play an important role in the establishment and maintenance of sex differences in the brain has garnered great enthusiasm but the reality in terms of actual advances has been slow. Two general approaches include the comparison of a particular epigenetic mark in males vs. females and the inhibition of key epigenetic enzymes or co-factors to determine if this eliminates a particular sex difference in brain or behavior. The majority of emphasis has been on candidate genes such as steroid receptors. Only recently have more generalized survey type approaches been achieved and these promise to open new vistas and accelerate discovery of important roles for DNA methylation, histone modification, genomic imprinting and microRNAs (miRs). Technical challenges abound and, while not unique to this field, will require novel thinking and new approaches by behavioral neuroendocrinologists.

16.
Front Neuroendocrinol ; 39: 28-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26368654

ABSTRACT

Fetal development could be considered a sensitive period wherein exogenous insults and changes to the maternal milieu can have long-term impacts on developmental programming. The placenta provides the fetus with protection and necessary nutrients for growth, and responds to maternal cues and changes in nutrient signaling through multiple epigenetic mechanisms. The X-linked enzyme O-linked-N-acetylglucosamine transferase (OGT) acts as a nutrient sensor that modifies numerous proteins to alter various cellular signals, including major epigenetic processes. This review describes epigenetic alterations in the placenta in response to insults during pregnancy, the potential links of OGT as a nutrient sensor to placental epigenetics, and the implications of placental epigenetics in long-term neurodevelopmental programming. We describe the role of placental OGT in the sex-specific programming of hypothalamic-pituitary-adrenal (HPA) axis programming deficits by early prenatal stress as an example of how placental signaling can have long-term effects on neurodevelopment.


Subject(s)
Epigenesis, Genetic/physiology , Fetal Development/genetics , Fetal Development/physiology , Fetus/metabolism , Placenta/physiology , Adult , Animals , Female , Humans , Pregnancy
17.
Nat Neurosci ; 18(5): 690-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25821913

ABSTRACT

The developing mammalian brain is destined for a female phenotype unless exposed to gonadal hormones during a perinatal sensitive period. It has been assumed that the undifferentiated brain is masculinized by direct induction of transcription by ligand-activated nuclear steroid receptors. We found that a primary effect of gonadal steroids in the highly sexually dimorphic preoptic area (POA) is to reduce activity of DNA methyltransferase (Dnmt) enzymes, thereby decreasing DNA methylation and releasing masculinizing genes from epigenetic repression. Pharmacological inhibition of Dnmts mimicked gonadal steroids, resulting in masculinized neuronal markers and male sexual behavior in female rats. Conditional knockout of the de novo Dnmt isoform, Dnmt3a, also masculinized sexual behavior in female mice. RNA sequencing revealed gene and isoform variants modulated by methylation that may underlie the divergent reproductive behaviors of males versus females. Our data show that brain feminization is maintained by the active suppression of masculinization via DNA methylation.


Subject(s)
Brain/growth & development , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , DNA, Intergenic/genetics , Disorders of Sex Development/genetics , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Preoptic Area/physiopathology , Sex Characteristics , Sex Differentiation/physiology , Animals , Copulation/drug effects , Copulation/physiology , CpG Islands , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Disorders of Sex Development/physiopathology , Estradiol/physiology , Female , Male , Mice , Microfilament Proteins/analysis , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phthalimides/pharmacology , Preoptic Area/enzymology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Rats , Rats, Sprague-Dawley , Testosterone/pharmacology , Testosterone/physiology , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
18.
J Neurosci ; 33(7): 2761-72, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23407936

ABSTRACT

Brain sexual differentiation in rodents results from the perinatal testicular androgen surge. In the preoptic area (POA), estradiol aromatized from testosterone upregulates the production of the proinflammatory molecule, prostaglandin E(2) (PGE(2)) to produce sex-specific brain development. PGE(2) produces a two-fold greater density of dendritic spines in males than in females and masculinizes adult copulatory behavior. One neonatal dose of PGE(2) masculinizes the POA and behavior, and simultaneous treatment with an inhibitor of additional prostaglandin synthesis prevents this masculinization, indicating a positive feedforward process that leads to sustained increases in PGE(2). The mechanisms underlying this feedforward process were unknown. Microglia, the primary immunocompetent cells in the brain, are active neonatally, contribute to normal brain development, and both produce and respond to prostaglandins. We investigated whether there are sex differences in microglia in the POA and whether they influence developmental masculinization. Neonatal males had twice as many ameboid microglia as females and a more activated morphological profile, and both estradiol and PGE(2) masculinized microglial number and morphology in females. Microglial inhibition during the critical period for sexual differentiation prevented sex differences in microglia, estradiol-induced masculinization of dendritic spine density, and adult copulatory behavior. Microglial inhibition also prevented the estradiol-induced upregulation of PGE(2), indicating that microglia are essential to the feedforward process through which estradiol upregulates prostaglandin production. These studies demonstrate that immune cells in the brain interact with the nervous and endocrine systems during development, and are crucial for sexual differentiation of brain and behavior.


Subject(s)
Behavior, Animal/physiology , Brain/cytology , Brain/physiology , Microglia/physiology , Sex Differentiation/physiology , Animals , Blotting, Western , Brain Chemistry/physiology , Cell Count , Cells, Cultured , Dendritic Spines/physiology , Dinoprostone/metabolism , Dinoprostone/physiology , Estradiol/pharmacology , Estradiol/physiology , Female , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Minocycline/pharmacology , Nerve Tissue Proteins/biosynthesis , Preoptic Area/growth & development , Preoptic Area/metabolism , Preoptic Area/physiology , Rats , Sexual Behavior, Animal/physiology , Sexual Maturation
19.
Front Neurosci ; 6: 26, 2012.
Article in English | MEDLINE | ID: mdl-22363256

ABSTRACT

Steroid hormones of gonadal origin act on the neonatal brain to produce sex differences that underlie adult reproductive physiology and behavior. Neuronal sex differences occur on a variety of levels, including differences in regional volume and/or cell number, morphology, physiology, molecular signaling, and gene expression. In the rodent, many of these sex differences are determined by steroid hormones, particularly estradiol, and are established by diverse downstream effects. One brain region that is potently organized by estradiol is the preoptic area (POA), a region critically involved in many behaviors that show sex differences, including copulatory and maternal behaviors. This review focuses on the POA as a case study exemplifying the depth and breadth of our knowledge as well as the gaps in understanding the mechanisms through which gonadal hormones produce lasting neural and behavioral sex differences. In the POA, multiple cell types, including neurons, astrocytes, and microglia are masculinized by estradiol. Multiple downstream molecular mediators are involved, including prostaglandins, various glutamate receptors, protein kinase A, and several immune signaling molecules. Moreover, emerging evidence indicates epigenetic mechanisms maintain sex differences in the POA that are organized perinatally and thereby produce permanent behavioral changes. We also review emerging strategies to better elucidate the mechanisms through which genetics and epigenetics contribute to brain and behavioral sex differences.

20.
J Neurosci ; 32(2): 593-8, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22238094

ABSTRACT

In immature neurons the amino acid neurotransmitter, GABA provides the dominant mode for neuronal excitation by inducing membrane depolarization due to Cl(-) efflux through GABA(A) receptors (GABA(A)Rs). The driving force for Cl(-) is outward because the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) elevates the Cl(-) concentration in these cells. GABA-induced membrane depolarization and the resulting activation of voltage-gated Ca(2+) channels is fundamental to normal brain development, yet the mechanisms that regulate depolarizing GABA are not well understood. The neurosteroid estradiol potently augments depolarizing GABA action in the immature hypothalamus by enhancing the activity of the NKCC1 cotransporter. Understanding how estradiol controls NKCC1 activity will be essential for a complete understanding of brain development. We now report that estradiol treatment of newborn rat pups significantly increases protein levels of two kinases upstream of the NKCC1 cotransporter, SPAK (STE20/SPS1-related proline alanine rich kinase) and OSR1 (oxidative stress response kinase). The estradiol-induced increase is transcription dependent, and its time course parallels that of estradiol-enhanced phosphorylation of NKCC1. Antisense oligonucleotide-mediated knockdown of SPAK, and to a lesser degree of OSR1, precludes estradiol-mediated enhancement of NKCC1 phosphorylation. Functionally, knockdown of SPAK or OSR1 in embryonic hypothalamic cultures diminishes estradiol-enhanced Ca(2+) influx induced by GABA(A)R activation. Our data suggest that SPAK and OSR1 may be critical factors in the regulation of depolarizing GABA-mediated processes in the developing brain. It will be important to examine these kinases with respect to sex differences and developmental brain anomalies in future studies.


Subject(s)
Estradiol/metabolism , Hypothalamus/growth & development , Hypothalamus/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Chloride Symporters/physiology , Animals , Animals, Newborn , Cells, Cultured , Estradiol/pharmacology , Female , Hypothalamus/cytology , Male , Phosphorylation/drug effects , Phosphorylation/physiology , Pregnancy , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Sex Characteristics , Solute Carrier Family 12, Member 2 , Up-Regulation/drug effects , Up-Regulation/physiology , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...