Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Environ Res ; 171: 177-184, 2019 04.
Article in English | MEDLINE | ID: mdl-30665119

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is suspected to have environmental and genetic contributions. Polychlorinated biphenyls (PCBs) are environmental risk factors of interest due to their potential as neurodevelopmental toxicants and environmental persistence despite a US production ban in the 1970s. METHODS: Participants were mother-child pairs from MARBLES, a high-risk pregnancy cohort that enrolls families who have one child diagnosed with ASD and are planning to have another child. PCB concentrations were measured in maternal blood at each trimester of pregnancy using gas chromatography coupled with triple quadruple mass spectrometry. Concentrations were summed into total PCB and two categories based on function/mechanisms of action: dioxin-like (DL), and ryanodine receptor (RyR)-activating PCBs. Multinomial logistic regression assessed risk of clinical outcome classification of ASD and non-typical development (Non-TD) compared to typically developing (TD) in the children at 3 years old. RESULTS: A total of 104 mother-child pairs were included. There were no significant associations for total PCB; however, there were borderline significant associations between DL-PCBs and decreased risk for Non-TD outcome classification (adjusted OR: 0.41 (95% CI 0.15-1.14)) and between RyR-activating PCBs and increased risk for ASD outcome classification (adjusted OR: 2.63 (95% CI 0.87-7.97)). CONCLUSION: This study does not provide strong supporting evidence that PCBs are risk factors for ASD or Non-TD. However, these analyses suggest the need to explore more deeply into subsets of PCBs as risk factors based on their function and structure in larger cohort studies where non-monotonic dose-response patterns can be better evaluated.


Subject(s)
Autism Spectrum Disorder/epidemiology , Environmental Exposure/statistics & numerical data , Environmental Pollutants , Polychlorinated Biphenyls , Calcium Carbonate , Child , Child, Preschool , Cohort Studies , Female , Gas Chromatography-Mass Spectrometry , Humans , Maternal Exposure , Pregnancy
2.
Arch Toxicol ; 92(11): 3337-3345, 2018 11.
Article in English | MEDLINE | ID: mdl-30225637

ABSTRACT

PCB 11 (3,3'-dichlorobiphenyl), a contemporary congener produced as a byproduct of current pigment production processes, has recently emerged as a prevalent worldwide pollutant. We recently demonstrated that exposure to PCB 11 increases dendritic arborization in vitro, but the mechanism(s) mediating this effect remain unknown. To address this data gap, primary cortical neuron-glia co-cultures derived from neonatal Sprague-Dawley rats were exposed for 48 h to either vehicle (0.1% DMSO) or PCB 11 at concentrations ranging from 1 fM to 1 nM in the absence or presence of pharmacologic antagonists of established molecular targets of higher chlorinated PCBs. Reporter cell lines were used to test activity of PCB 11 at the aryl hydrocarbon receptor (AhR) and thyroid hormone receptor (THR). PCB 11 lacked activity at the AhR and THR, and antagonism of these receptors had no effect on the dendrite-promoting activity of PCB 11. Pharmacologic antagonism of various calcium channels or treatment with antioxidants also did not alter PCB 11-induced dendritic arborization. In contrast, pharmacologic blockade or shRNA knockdown of cAMP response element-binding protein (CREB) significantly decreased dendritic growth in PCB 11-exposed cultures, suggesting PCB 11 promotes dendritic growth via CREB-mediated mechanisms. Since CREB signaling is crucial for normal neurodevelopment, and perturbations of CREB signaling have been associated with neurodevelopmental disorders, our findings suggest that this contemporary pollutant poses a threat to the developing brain, particularly in individuals with heritable mutations that promote CREB signaling.


Subject(s)
Cerebral Cortex/drug effects , Cyclic AMP Response Element-Binding Protein/physiology , Dendrites/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Calcium/metabolism , Cells, Cultured , Dendrites/physiology , Humans , Mice , Neuroglia/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/physiology
3.
Arch Toxicol ; 92(10): 3163-3173, 2018 10.
Article in English | MEDLINE | ID: mdl-30132043

ABSTRACT

Polychlorinated biphenyls (PCBs), and in particular non-dioxin-like (NDL) congeners, continue to pose a significant risk to the developing nervous system. PCB 95, a prevalent NDL congener in the human chemosphere, promotes dendritic growth in rodent primary neurons by activating calcium-dependent transcriptional mechanisms that normally function to link activity to dendritic growth. Activity-dependent dendritic growth is also mediated by calcium-dependent translational mechanisms involving mechanistic target of rapamycin (mTOR), suggesting that the dendrite-promoting activity of PCB 95 may also involve mTOR signaling. Here, we test this hypothesis using primary neuron-glia co-cultures derived from the hippocampi of postnatal day 0 Sprague Dawley rats. PCB 95 (1 nM) activated mTOR in hippocampal cultures as evidenced by increased phosphorylation of mTOR at ser2448. Pharmacologic inhibition of mTOR signaling using rapamycin (20 nM), FK506 (5 nM), or 4EGI-1 (1 µM), and siRNA knockdown of mTOR, or the mTOR complex binding proteins, raptor or rictor, blocked PCB 95-induced dendritic growth. These data identify mTOR activation as a novel molecular mechanism contributing to the effects of PCB 95 on dendritic arborization. In light of clinical data linking gain-of-function mutations in mTOR signaling to neurodevelopmental disorders, our findings suggest that mTOR signaling may represent a convergence point for gene by environment interactions that confer risk for adverse neurodevelopmental outcomes.


Subject(s)
Dendrites/drug effects , Hippocampus/cytology , Neurons/drug effects , Polychlorinated Biphenyls/toxicity , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Dendrites/physiology , Female , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Neuroglia/cytology , Neurons/metabolism , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Protein 1A/metabolism
4.
Differentiation ; 93: 66-71, 2017.
Article in English | MEDLINE | ID: mdl-27918915

ABSTRACT

Androgen, beta-catenin (CTNNB1), and estrogen pathways stimulate proliferative growth of developing mouse prostate but how these pathways interact is not fully understood. We previously found that androgens induce CTNNB1 signaling in mouse urogenital sinus (UGS) epithelium from which prostatic ductal epithelium derives. Others have shown that low estradiol concentrations induce UGS epithelial proliferative growth. Here, we found that CTNNB1 signaling overlaps cyclin D1 (CCND1) expression in prostatic buds and we used a genetic approach to test whether CTNNB1 signaling induces CCND1 expression. We observed an unexpected sexually dimorphic response to hyperactive CCNTB1 signaling: in male mouse UGS it increased Ccnd1 mRNA abundance without increasing its protein abundance but in female UGS it increased Ccnd1 mRNA and protein abundance, suggesting a potential role for estrogens in stabilizing CCND1 protein. Treating wild type male UGS explants with androgen and either 17ß-estradiol or a proteasome inhibitor increased CCND1 protein and KI67 labeling in prostatic bud epithelium. Together, our results are consistent with an epithelial proliferative growth mechanism linking CTNNB1-driven Ccnd1 transcription and estrogen-mediated CCND1 protein stabilization.


Subject(s)
Cyclin D1/genetics , Embryonic Development/genetics , Estrogens/genetics , beta Catenin/genetics , Androgens/genetics , Animals , Epithelium/growth & development , Epithelium/metabolism , Estrogens/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Organogenesis , Prostate , RNA, Messenger/genetics , beta Catenin/metabolism
5.
Neurourol Urodyn ; 35(2): 192-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25394276

ABSTRACT

AIMS: Mice are increasingly being used as models to investigate aspects of urinary dysfunction that humans with lower urinary tract symptoms (LUTS) experience. One method used to examine voiding function is the spontaneous void spot assay. The purpose of this study was to characterize and identify animal husbandry conditions that might confound results of the spontaneous void spot assay in male C57Bl/6J mice. METHODS: Mice were placed in cages lined with filter paper for 4 hr and urine was visualized with UV transillumination. Voiding parameters including urine spot number, spot size, total urine area, primary void area, corner and center voiding were quantified. RESULTS: Adult male mice void more frequently with advancing age and a subpopulation (5-10%) display a frequent spotting pattern at 6-9 weeks of age. Voiding was not significantly different in male mice weaned to group housing (4-6 per cage) versus single housing, and was not altered when they were used as breeders. Voiding was changed upon transferring group housed adult males to single density cages, which decreased total urine area. Repeated assays of male voiding behavior over three consecutive days increased primary void area by the third day of monitoring and revealed that voiding behavior is impacted by routine cage changes and time of day. CONCLUSIONS: Together these results identify housing and husbandry practices that influence male voiding behaviors in the spontaneous void spot assay and will inform voiding behavior analyses conducted with male C57Bl/6J mice.


Subject(s)
Animal Husbandry/methods , Diagnostic Techniques, Urological , Housing, Animal , Urination , Urodynamics , Age Factors , Animals , Behavior, Animal , Circadian Rhythm , Handling, Psychological , Male , Mice, Inbred C57BL , Predictive Value of Tests , Reproducibility of Results , Time Factors
6.
Dev Dyn ; 244(11): 1404-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26283270

ABSTRACT

BACKGROUND: Epigenetic factors influence stem cell function and other developmental events but their role in prostate morphogenesis is not completely known. We tested the hypothesis that histone deacetylase (HDAC) activity is required for prostate morphogenesis. RESULTS: We identified the presence of class I nuclear HDACs in the mouse urogenital sinus (UGS) during prostate development and found that Hdac 2 mRNA abundance diminishes as development proceeds which is especially evident in prostatic epithelium. Blockade of HDACs with the inhibitor trichostatin A (TSA) decreased the number of prostatic buds formed in UGS explant cultures but not the number of buds undergoing branching morphogenesis. In the latter, TSA promoted an extensive branching phenotype that was reversed by exogenous NOGGIN protein, which functions as a bone morphogenetic protein (BMP) inhibitor. TSA also increased Bmp2 promoter H3K27ac abundance, Bmp2 and Bmp4 mRNA abundance, and the percentage of epithelial cells marked by BMP-responsive phosphorylated SMAD1/5/8 protein. TSA exposed UGS explants grafted under the kidney capsule of untreated host mice for continued development achieved a smaller size without an obvious difference in glandular histology compared with control treated grafts. CONCLUSIONS: These results are consistent with an active role for HDACs in shaping prostate morphogenesis by regulating Bmp abundance.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Histones/metabolism , Prostate/growth & development , Acetylation , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/metabolism , Chromatin/metabolism , Chromatin Immunoprecipitation , Epigenesis, Genetic , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylase 2/metabolism , Humans , Hydroxamic Acids/chemistry , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Morphogenesis , Phenotype , Promoter Regions, Genetic , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Signal Transduction
7.
Dev Biol ; 387(2): 142-53, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24503032

ABSTRACT

In prostate and other epithelial cancers, E-cadherin (CDH1) is downregulated inappropriately by DNA methylation to promote an invasive phenotype. Though cancer frequently involves a reawakening of developmental signaling pathways, whether DNA methylation of Cdh1 occurs during organogenesis has not been determined. Here we show that DNA methylation of Cdh1 mediates outgrowth of developing prostate ducts. During the three-day gestational window leading up to and including prostate ductal initiation, Cdh1 promoter methylation increases and its mRNA and protein abundance decreases in epithelium giving rise to prostatic buds. DNA methylation is required for prostate specification, ductal outgrowth, and branching morphogenesis. All three endpoints are impaired by a DNA methylation inhibitor, which also decreases Cdh1 promoter methylation and increases Cdh1 mRNA and protein abundance. A CDH1 function-blocking antibody restores prostatic identity, bud outgrowth, and potentiates epithelial differentiation in the presence of the DNA methylation inhibitor. This is the first study to mechanistically link acquired changes in DNA methylation to the normal process of prostate organogenesis. We propose a novel mechanism whereby Cdh1 promoter methylation restricts Cdh1 abundance in developing prostate epithelium to create a permissive environment for prostatic bud outgrowth. Thus, DNA methylation primes the prostate primordium to respond to developmental cues mediating outgrowth, differentiation and maturation of the ductal network.


Subject(s)
Cadherins/genetics , Cdh1 Proteins/metabolism , DNA Methylation/genetics , Gene Expression Regulation, Developmental , Prostate/embryology , Animals , Antibodies, Blocking/immunology , Cdh1 Proteins/genetics , Cdh1 Proteins/immunology , Cell Differentiation/immunology , Epithelium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Morphogenesis/genetics , Promoter Regions, Genetic/genetics , Prostate/metabolism , RNA, Messenger/metabolism
8.
Dev Biol ; 396(2): 237-45, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25446526

ABSTRACT

Androgen receptor (AR) signaling initiates mouse prostate development by stimulating prostate ductal bud formation and specifying bud patterns. Curiously, however, prostatic bud initiation lags behind the onset of gonadal testosterone synthesis by about three days. This study's objective was to test the hypothesis that DNA methylation controls the timing and scope of prostate ductal development by regulating Ar expression in the urogenital sinus (UGS) from which the prostate derives. We determined that Ar DNA methylation decreases in UGS mesenchyme during prostate bud formation in vivo and that this change correlates with decreased DNA methyltransferase expression in the same cell population during the same time period. To examine the role of DNA methylation in prostate development, fetal UGSs were grown in serum-free medium and 5 alpha dihydrotestosterone (DHT) and the DNA methylation inhibitor 5'-aza-2'-deoxycytidine (5AzadC) were introduced into the medium at specific times. As a measure of prostate development, in situ hybridization was used to visualize and count Nkx3-1 mRNA positive prostatic buds. We determined that inhibiting DNA methylation when prostatic buds are being specified, accelerates the onset of prostatic bud development, increases bud number, and sensitizes the budding response to androgens. Inhibition of DNA methylation also reduces Ar DNA methylation in UGS explants and increases Ar mRNA and protein in UGS mesenchyme and epithelium. Together, these results support a novel mechanism whereby Ar DNA methylation regulates UGS androgen sensitivity to control the rate and number of prostatic buds formed, thereby establishing a developmental checkpoint.


Subject(s)
DNA Methylation/physiology , Gene Expression Regulation, Developmental/physiology , Models, Biological , Organogenesis/physiology , Prostate/embryology , Receptors, Androgen/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Azacitidine/analogs & derivatives , Base Sequence , DNA Primers/genetics , Decitabine , Dihydrotestosterone , Homeodomain Proteins/metabolism , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , In Vitro Techniques , Male , Mice , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Sequence Analysis, DNA , Transcription Factors/metabolism
9.
Am J Physiol Renal Physiol ; 308(12): F1369-78, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25904700

ABSTRACT

Void spot assays (VSA) and cystometry are two of the most common tests performed in mice to assess lower urinary tract function. Assay protocols and methodology vary greatly among laboratories, and little is known about reproducibility of results generated by different laboratories. We performed VSA in four mouse strains, comparing males with females and comparing results between two independent laboratories. Unique aspects of the current study include direct comparison of results of VSA performed in a similar manner in two locations and comparison of cystometry performed using two different rates of infusion in these two laboratories. Both assays were performed in male and female 129S1/SvImJ, C57BL/6J, NOD/ShiLtJ, and CAST/EiJ mice, and cystometry was performed under urethane anesthesia (10/group). Assays were performed and results analyzed as previously described. Results obtained in female mice were compared with previously reported values. Results of lower urinary tract function testing in mice vary in a consistent manner with strain and sex. Variables in husbandry, testing techniques, and analysis of results can significantly affect conclusions, particularly those obtained by cystometry. Although VSA results were remarkably similar between the two laboratories, consistent methods for performing lower urinary tract function testing in mice are required to compare results among studies with confidence.


Subject(s)
Urethane/analysis , Urinary Bladder/physiology , Urination/genetics , Urodynamics/genetics , Animals , Female , Male , Mice , Mice, Inbred NOD , Reproducibility of Results , Sex Factors , Urination/physiology , Urodynamics/physiology
10.
Am J Physiol Renal Physiol ; 308(12): F1431-43, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25855514

ABSTRACT

Aging men are susceptible to developing lower urinary tract symptoms, but the underlying etiology is unknown and the influence of dietary and environmental factors on them is unclear. We tested whether a folic acid-enriched diet changed urinary tract physiology and biology in control male mice and male mice with urinary dysfunction induced by exogenous testosterone and estradiol (T+E2), which mimics changing hormone levels in aging humans. T+E2 treatment increased mouse urine output, time between voiding events, and bladder capacity and compliance. Consumption of a folic acid-enriched diet moderated these changes without decreasing prostate wet weight or threshold voiding pressure. One potential mechanism for these changes involves water balance. T+E2 treatment increases plasma concentrations of anti-diuretic hormone, which is offset at least in part by a folic acid-enriched diet. Another potential mechanism involves neural control of micturition. The folic acid-enriched diet, fed to T+E2-treated mice, increased voiding frequency in response to intravesicular capsaicin infusion and increased mRNA abundance of the capsaicin-sensitive cation channel transient receptor potential vanilloid subfamily member 1 (Trpv1) in L6 and S1 dorsal root ganglia (DRG) neurons. T+E2 treatment and a folic acid-enriched diet also modified DNA methylation, which is capable of altering gene expression. We found the enriched diet increased global DNA methylation in dorsal and ventral prostate and L6 and S1 DRG. Our results are consistent with folic acid acting to slow or reverse T+E2-mediated alteration in urinary function in part by normalizing water balance and enhancing or preserving afferent neuronal function.


Subject(s)
Estradiol/pharmacology , Folic Acid/pharmacology , Lower Urinary Tract Symptoms/drug therapy , Testosterone/pharmacology , Urinary Tract Physiological Phenomena/drug effects , Animal Feed , Animals , Capsaicin/administration & dosage , Capsaicin/pharmacology , Diet , Estradiol/administration & dosage , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Mice, Inbred C57BL , Testosterone/administration & dosage , Urinary Bladder/drug effects , Urinary Bladder/metabolism
11.
J Urol ; 193(2): 722-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25167991

ABSTRACT

PURPOSE: Estrogens are important in prostate growth and have a role in benign prostatic hyperplasia. However, to our knowledge no current therapy directly targets estrogen action. Estrogens act primarily via estrogen receptors α and ß. In a mouse model we evaluated the relative contribution of these receptors to bladder complications of benign prostatic hyperplasia. We also evaluated the prevention of these bladder complications using the selective estrogen receptor modulators raloxifene and tamoxifen (estrogen receptor-α selective antagonists), and R,R-THC (estrogen receptor-ß selective antagonist). MATERIALS AND METHODS: Adult male C57bl/6 mice received implants of 25 mg testosterone and 2.5 mg 17ß-estradiol slow release pellets. Untreated controls underwent sham surgery. We evaluated the contributions of the estrogen receptor subtypes in ERαKO and ERßKO mice compared to their respective wild-type litter mates. Wild-type mice treated with testosterone plus 17ß-estradiol were compared to mice treated with testosterone plus 17ß-estradiol and 25 mg selective estrogen receptor modulators to evaluate the prevention of benign prostatic hyperplasia complications by selective estrogen receptor modulators. RESULTS: Large bladders with urinary retention developed in ERαWT and ERßWT litter mates treated with testosterone plus 17ß-estradiol but such bladders did not develop in ERαKO mice treated with testosterone plus 17ß-estradiol. ERßKO mice treated with testosterone plus 17ß-estradiol had large bladders with urinary retention and increased bladder mass. Cotreatment with the estrogen receptor-α antagonist raloxifene resulted in decreased bladder mass compared to that in wild-type mice treated with testosterone plus 17ß-estradiol. Bladders in mice treated with the estrogen receptor-ß antagonist R,R-THC were similar to those in testosterone plus 17ß-estradiol treated mice. CONCLUSIONS: Estrogen receptor-α but not ß is a key mediator of bladder complications of benign prostatic hyperplasia and a potential target for future therapies.


Subject(s)
Estrogen Receptor alpha , Estrogen Receptor beta , Prostatic Hyperplasia/complications , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Urinary Bladder Diseases , Animals , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/physiology , Male , Mice , Mice, Inbred C57BL , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Urinary Bladder Diseases/drug therapy , Urinary Bladder Diseases/etiology
12.
Dev Biol ; 376(2): 125-35, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23396188

ABSTRACT

Fetal prostate development is initiated by androgens and patterned by androgen dependent and independent signals. How these signals integrate to control epithelial cell differentiation and prostatic bud patterning is not fully understood. To test the role of beta-catenin (Ctnnb1) in this process, we used a genetic approach to conditionally delete or stabilize Ctnnb1 in urogenital sinus (UGS) epithelium from which the prostate derives. Two opposing mechanisms of action were revealed. By deleting Ctnnb1, we found it is required for separation of UGS from cloaca, emergence or maintenance of differentiated UGS basal epithelium and formation of prostatic buds. By genetically inducing a patchy subset of UGS epithelial cells to express excess CTNNB1, we found its excess abundance increases Bmp expression and leads to a global impairment of prostatic bud formation. Addition of NOGGIN partially restores prostatic budding in UGS explants with excess Ctnnb1. These results indicate a requirement for Ctnnb1 in UGS basal epithelial cell differentiation, prostatic bud initiation and bud spacing and suggest some of these actions are mediated in part through activation of BMP signaling.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 7/metabolism , Gene Expression Regulation, Developmental , beta Catenin/biosynthesis , Animals , Body Patterning , Cell Differentiation , Crosses, Genetic , Epithelial Cells/cytology , Gene Deletion , Male , Mice , Microscopy, Electron, Scanning/methods , Models, Genetic , Prostate/embryology , Signal Transduction
13.
Differentiation ; 84(3): 232-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22898663

ABSTRACT

The purpose of this study was to validate a combined in situ hybridization (ISH)/immunohistochemistry (IHC) staining method for visualizing and quantifying mouse prostatic buds. To refine animal usage in prostate development studies, we also determined whether a comparable number of prostatic buds were formed in male and female mouse urogenital sinus (UGS) explants grown in vitro in the presence of androgen. We used IHC to label UGS epithelium and ISH to label prostatic buds with one of three different prostatic bud marking riboprobes: a previously identified prostatic bud marker, NK-3 transcription factor, locus 1 (Nkx3-1), and two newly identified prostatic bud markers, wingless-related MMTV integration site 10b (Wnt10b) and ectodysplasin-A receptor (Edar). We calculated total buds formed per UGS and the proportion marked by each mRNA after male UGS development in vivo and male and female UGS development in vitro. Nkx3-1 was first to mark the prostate field during UGS development in vivo but all three mRNAs marked prostatic buds during later developmental stages. The mRNAs localized to different domains: Nkx3-1 was present along about half the prostatic bud length while Edar and Wnt10b were restricted to distal bud tips. None of the mRNAs marked all buds formed in vitro and the proportion marked was developmental stage- and gender-dependent. Nkx3-1 marked the highest proportion of prostatic buds during in vitro UGS development. Together, our results reveal that ISH staining of mouse UGS can be used to quantify prostatic bud number, Nkx3-1 is currently the best suited riboprobe for this method, and female UGSs cannot be used interchangeably with male UGSs when conducting prostate development studies in vitro. We also found that Nkx3-1, Edar, and Wnt10b mark different prostatic bud regions and are likely to be useful in future studies of regional differences in prostatic bud gene expression.


Subject(s)
In Situ Hybridization/methods , Prostate/embryology , Animals , Edar Receptor/genetics , Edar Receptor/metabolism , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Prostate/metabolism , RNA, Messenger/biosynthesis , Sex Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
14.
Dev Dyn ; 241(12): 2005-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23074159

ABSTRACT

BACKGROUND: Prostate morphogenesis initiates in the urogenital sinus (UGS) with epithelial bud development. Sulfatase-1 (SULF1) inhibits bud development by reducing extracellular heparan sulfate (HS) 6-O sulfation and impairing FGF10 signaling by means of the ERK1/2 mitogen activated kinases. RESULTS: We characterized the expression patterns of HS 6-O sulfation modifying enzymes in the developing prostate by in situ hybridization and showed that Sulf1 and Hs6st1 had overlapping but distinct expression domains. Notably, Hs6st1 was present while Sulf1 was excluded from the tips of elongating epithelial buds. This predicted relatively high HS 6-O sulfation at the tips of elongating epithelial buds that was confirmed by immunohistochemistry. The pattern of Sulf1 expression in the peri-mesenchymal epithelium matched predicted locations of bone morphogenetic protein (BMP) signaling. Exogenous BMP4 and BMP7 induced Sulf1 expression in the UGS, decreased epithelial HS 6-O sulfation, and reduced ERK1/2 activation in response to FGF10. CONCLUSIONS: These data suggest that BMPs limit FGF10 action in the developing prostate at least in part by inducing Sulf1.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Heparitin Sulfate/metabolism , Prostate/embryology , Sulfotransferases/biosynthesis , Animals , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Female , Fibroblast Growth Factor 10/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heparitin Sulfate/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Prostate/cytology , Sulfotransferases/genetics
15.
Dev Dyn ; 240(11): 2548-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21936019

ABSTRACT

Prostate development is influenced by ß-catenin signaling, but it is unclear which ß-catenin activators are involved, where they are synthesized, and whether their mRNA abundance is influenced by androgens. We identified WNT/ß-catenin-responsive ß-galactosidase activity in the lower urogenital tract (LUT) of transgenic reporter mice, but ß-galactosidase activity differed among the four mouse strains we examined. We used in situ hybridization to compare patterns of Wnts, r-spondins (Rspos, co-activators of ß-catenin signaling), ß-catenin-responsive mRNAs, and an androgen receptor-responsive mRNA in wild type fetal male, fetal female, and neonatal male LUT. Most Wnt and Rspo mRNAs were present in LUT during prostate development. Sexually dimorphic expression patterns were observed for WNT/ß-catenin-responsive genes, and for Wnt2b, Wnt4, Wnt7a, Wnt9b, Wnt10b, Wnt11, Wnt16, and Rspo3 mRNAs. These results reveal sexual differences in WNT/ß-catenin signaling in fetal LUT, supporting the idea that this pathway may be directly or indirectly responsive to androgens during prostate ductal development.


Subject(s)
Thrombospondins/genetics , Urogenital System/embryology , Urogenital System/metabolism , Wnt Proteins/genetics , Anatomy, Artistic , Animals , Animals, Newborn , Atlases as Topic , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thrombospondins/metabolism , Tissue Distribution , Urogenital System/growth & development , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
16.
Dev Dyn ; 240(10): 2364-77, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21905163

ABSTRACT

Epithelial-stromal interactions in the lower urogenital tract (LUT) are integral to prostatic and seminal vesicle development in males, vaginal and uterine development in females, and urethral development in both sexes. Gene expression profiling of isolated LUT stroma and epithelium has unraveled mechanisms of LUT development, but such studies are confounded by heterogeneous and ill-defined cell sub-populations contained within each tissue compartment. We used in situ hybridization to synthesize a high-resolution molecular atlas of 17-day post-coitus fetal mouse LUT. We identified mRNAs that mark selective cell populations of the seminal vesicle, ejaculatory duct, prostate, urethra, and vagina, subdividing these tissues into 16 stromal and 8 epithelial sub-compartments. These results provide a powerful tool for mapping LUT gene expression patterns and also reveal previously uncharacterized sub-compartments that may play mechanistic roles in LUT development of which we were previously unaware.


Subject(s)
Biomarkers/metabolism , Urogenital System/anatomy & histology , Urogenital System/embryology , Urogenital System/metabolism , Animals , Atlases as Topic , Female , Gene Expression Profiling , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
17.
J Neurosci Methods ; 341: 108793, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32461071

ABSTRACT

BACKGROUND: Primary neuronal cell cultures are useful for studying mechanisms that influence dendritic morphology during normal development and in response to various stressors. However, analyzing dendritic morphology is challenging, particularly in cultures with high cell density, and manual methods of selecting neurons and tracing dendritic arbors can introduce significant bias, and are labor-intensive. To overcome these challenges, semi-automated and automated methods are being developed, with most software solutions requiring computer-assisted dendrite tracing with subsequent quantification of various parameters of dendritic morphology, such as Sholl analysis. However fully automated approaches for classic Sholl analysis of dendritic complexity are not currently available. NEW METHOD: The previously described Omnisphero software, was extended by adding new functions to automatically assess dendritic mass, total length of the dendritic arbor and the number of primary dendrites, branch points, and terminal tips, and to perform Sholl analysis. RESULTS: The new functions for assessing dendritic morphology were validated using primary mouse hippocampal and rat cortical neurons transfected with a fluorescently tagged MAP2 cDNA construct. These functions allow users to select specific populations of neurons as a training set for subsequent automated selection of labeled neurons in high-density cultures. COMPARISON WITH EXISTING SEMI-AUTOMATED METHODS: Compared to manual or semi-automated analyses of dendritic arborization, the new functions increase throughput while significantly decreasing researcher bias associated with neuron selection, tracing, and thresholding. CONCLUSION: These results demonstrate the importance of using unbiased automated methods to mitigate experimenter-dependent bias in analyzing dendritic morphology.


Subject(s)
Hippocampus , Neurons , Animals , Dendrites , Image Processing, Computer-Assisted , Mice , Neuronal Plasticity , Rats
18.
Toxicol Sci ; 168(1): 95-109, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30395321

ABSTRACT

Early life exposures to environmental contaminants are implicated in the pathogenesis of many neurodevelopmental disorders (NDDs). These disorders often display sex biases, but whether environmental neurotoxicants act in a sex-dependent manner to modify neurodevelopment is largely unknown. Since altered dendritic morphology is associated with many NDDs, we tested the hypothesis that male and female primary mouse neurons are differentially susceptible to the dendrite-promoting activity of 2,2',3,5',6-pentachlorobiphenyl (PCB 95). Hippocampal and cortical neuron-glia co-cultures were exposed to vehicle (0.1% dimethylsulfoxide) or PCB 95 (100 fM-1 µM) from day in vitro 7-9. As determined by Sholl analysis, PCB 95-enhanced dendritic growth in female but not male hippocampal and cortical neurons. In contrast, both male and female neurons responded to bicuculline with increased dendritic complexity. Detailed morphometric analyses confirmed that PCB 95 effects on the number and length of primary and nonprimary dendrites varied depending on sex, brain region and PCB concentration, and that female neurons responded more consistently with increased dendritic growth and at lower concentrations of PCB 95 than their male counterparts. Exposure to PCB 95 did not alter cell viability or the ratio of neurons to glia in cultures of either sex. These results demonstrate that cultured female mouse hippocampal and cortical neurons are more sensitive than male neurons to the dendrite-promoting activity of PCB 95, and suggest that mechanisms underlying PCB 95-induced dendritic growth are sex-dependent. These data highlight the importance of sex in neuronal responses to environmental neurotoxicants.


Subject(s)
Dendrites/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Polychlorinated Biphenyls/pharmacology , Animals , Astrocytes/drug effects , Bicuculline/pharmacology , Cell Survival/drug effects , Cerebral Cortex/anatomy & histology , Cerebral Cortex/drug effects , Coculture Techniques , Female , Hippocampus/anatomy & histology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , Sex Factors
19.
Genes Brain Behav ; 18(1): e12526, 2019 01.
Article in English | MEDLINE | ID: mdl-30311737

ABSTRACT

Dendritic morphology is a critical determinant of neuronal connectivity, and calcium signaling plays a predominant role in shaping dendrites. Altered dendritic morphology and genetic mutations in calcium signaling are both associated with neurodevelopmental disorders (NDDs). In this study we tested the hypothesis that dendritic arborization and NDD-relevant behavioral phenotypes are altered by human mutations that modulate calcium-dependent signaling pathways implicated in NDDs. The dendritic morphology of pyramidal neurons in CA1 hippocampus and somatosensory cortex was quantified in Golgi-stained brain sections from juvenile mice of both sexes expressing either a human gain-of-function mutation in ryanodine receptor 1 (T4826I-RYR1), a human CGG repeat expansion (170-200 CGG repeats) in the fragile X mental retardation gene 1 (FMR1 premutation), both mutations (double mutation; DM), or wildtype mice. In hippocampal neurons, increased dendritic arborization was observed in male T4826I-RYR1 and, to a lesser extent, male FMR1 premutation neurons. Dendritic morphology of cortical neurons was altered in both sexes of FMR1 premutation and DM animals with the most pronounced differences seen in DM females. Genotype also impaired behavior, as assessed using the three-chambered social approach test. The most striking lack of sociability was observed in DM male and female mice. In conclusion, mutations that alter the fidelity of calcium signaling enhance dendritic arborization in a brain region- and sex-specific manner and impair social behavior in juvenile mice. The phenotypic outcomes of these mutations likely provide a susceptible biological substrate for additional environmental stressors that converge on calcium signaling to determine individual NDD risk.


Subject(s)
Calcium Signaling , Dendrites/metabolism , Gain of Function Mutation , Pyramidal Cells/cytology , Social Behavior , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Dendrites/physiology , Female , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, Inbred C57BL , Neuronal Outgrowth , Neuronal Plasticity , Pyramidal Cells/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Trinucleotide Repeat Expansion
20.
Biol Open ; 8(3)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30745437

ABSTRACT

Beta-catenin (CTNNB1) directs ectodermal appendage spacing by activating ectodysplasin A receptor (EDAR) transcription, but whether CTNNB1 acts by a similar mechanism in the prostate, an endoderm-derived tissue, is unclear. Here we examined the expression, function, and CTNNB1 dependence of the EDAR pathway during prostate development. In situ hybridization studies reveal EDAR pathway components including Wnt10b in the developing prostate and localize these factors to prostatic bud epithelium where CTNNB1 target genes are co-expressed. We used a genetic approach to ectopically activate CTNNB1 in developing mouse prostate and observed focal increases in Edar and Wnt10b mRNAs. We also used a genetic approach to test the prostatic consequences of activating or inhibiting Edar expression. Edar overexpression does not visibly alter prostatic bud formation or branching morphogenesis, and Edar expression is not necessary for either of these events. However, Edar overexpression is associated with an abnormally thick and collagen-rich stroma in adult mouse prostates. These results support CTNNB1 as a transcriptional activator of Edar and Wnt10b in the developing prostate and demonstrate Edar is not only important for ectodermal appendage patterning but also influences collagen organization in adult prostates.This article has an associated First Person interview with the first author of the paper.

SELECTION OF CITATIONS
SEARCH DETAIL