Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
1.
Cell ; 171(1): 242-255.e27, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938116

ABSTRACT

The morphogenesis of branched organs remains a subject of abiding interest. Although much is known about the underlying signaling pathways, it remains unclear how macroscopic features of branched organs, including their size, network topology, and spatial patterning, are encoded. Here, we show that, in mouse mammary gland, kidney, and human prostate, these features can be explained quantitatively within a single unifying framework of branching and annihilating random walks. Based on quantitative analyses of large-scale organ reconstructions and proliferation kinetics measurements, we propose that morphogenesis follows from the proliferative activity of equipotent tips that stochastically branch and randomly explore their environment but compete neutrally for space, becoming proliferatively inactive when in proximity with neighboring ducts. These results show that complex branched epithelial structures develop as a self-organized process, reliant upon a strikingly simple but generic rule, without recourse to a rigid and deterministic sequence of genetically programmed events.


Subject(s)
Kidney/growth & development , Mammary Glands, Human/growth & development , Models, Biological , Morphogenesis , Prostate/growth & development , Animals , Female , Humans , Kidney/embryology , Male , Mammary Glands, Human/embryology , Mice , Prostate/embryology
2.
Dev Biol ; 473: 50-58, 2021 05.
Article in English | MEDLINE | ID: mdl-33529704

ABSTRACT

The prostate develops by epithelial budding and branching processes that occur during fetal and postnatal stages. The adult prostate demonstrates remarkable regenerative capacity, with the ability to regrow to its original size over multiple cycles of castration and androgen administration. This capacity for controlled regeneration prompted the search for an androgen-independent epithelial progenitor in benign prostatic hyperplasia (BPH) and prostate cancer (PCa). BPH is hypothesized to be a reawakening of ductal branching, resulting in the formation of new proximal glands, all while androgen levels are decreasing in the aging male. Advanced prostate cancer can be slowed with androgen deprivation, but resistance eventually occurs, suggesting the existence of an androgen-independent progenitor. Recent studies indicate that there are multiple castration-insensitive epithelial cell types in the proximal area of the prostate, but not all act as progenitors during prostate development or regeneration. This review highlights how recent cellular and anatomical studies are changing our perspective on the identity of the prostate progenitor.


Subject(s)
Prostate/metabolism , Prostate/pathology , Stem Cells/metabolism , Androgen Antagonists/metabolism , Androgens/metabolism , Animals , Cell Differentiation , Epithelial Cells/metabolism , Humans , Male , Organogenesis , Prostate/embryology , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism
3.
Development ; 146(20)2019 10 23.
Article in English | MEDLINE | ID: mdl-31575645

ABSTRACT

The prostate is formed by a branched glandular epithelium composed of basal cells (BCs) and luminal cells (LCs). Multipotent and unipotent stem cells (SCs) mediate the initial steps of prostate development whereas BCs and LCs are self-sustained in adult mice by unipotent lineage-restricted SCs. The spatiotemporal regulation of SC fate and the switch from multipotency to unipotency remain poorly characterised. Here, by combining lineage tracing, whole-tissue imaging, clonal analysis and proliferation kinetics, we uncover the cellular dynamics that orchestrate prostate postnatal development in mouse. We found that at an early stage of development multipotent basal SCs are located throughout the epithelium and are progressively restricted at the distal tip of the ducts, where, together with their progeny, they establish the different branches and the final structure of prostate. In contrast, pubertal development is mediated by unipotent lineage-restricted SCs. Our results uncover the spatiotemporal regulation of the switch from multipotency to unipotency during prostate development.


Subject(s)
Multipotent Stem Cells/cytology , Prostate/cytology , Prostate/embryology , Animals , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation/physiology , Cells, Cultured , Male , Mice , Multipotent Stem Cells/metabolism , Organogenesis/physiology , Prostate/metabolism
4.
Dev Dyn ; 250(5): 618-628, 2021 05.
Article in English | MEDLINE | ID: mdl-33325097

ABSTRACT

Several studies reported the concerted and mutual communication between the prostate epithelium and stroma, which determines the final organ architecture and function, but gets awry in cancer. Deciphering the mechanisms involved in this communication is crucial to find new therapeutic strategies. HS sequesters a number of secreted growth factors and cytokines, controlling their bioavailability to the target cells, suggesting that HS is an important regulator of the extracellular matrix (ECM) and a key player in the cell-cell and cell-microenvironment communication during prostate morphogenesis and physiology. We propose that by controlling HS biosynthesis and sulfation pattern, as well as the cleavage of the HS chain and/or the shedding of proteoglycans, epithelial and stromal cells are able to precisely tune the availability of signaling molecules and modulate ligand-receptor interaction and intracellular signal transduction.


Subject(s)
Heparitin Sulfate/biosynthesis , Prostate/metabolism , Animals , Glucuronidase/metabolism , Humans , Male , Prostate/embryology , Signal Transduction
5.
Prostate ; 81(3): 214-219, 2021 02.
Article in English | MEDLINE | ID: mdl-33393689

ABSTRACT

BACKGROUND: The second gestational trimester is a very important period for male genital development. We analyzed the testicular and prostatic volume growth and compared them to the biometric parameters in human fetuses. METHODS: We studied 64 testes and 32 prostates from 32 fetuses, aged 10-22 weeks postconception. Fetuses were evaluated regarding total length, crown-rump length, and bodyweight. The same observer performed all the measurements. After testicular and prostatic dissection, the prostate and testicular length, width and thickness were recorded with the aid of computer programs (Image Pro and ImageJ software, version 1.46r). Testicular volume (TV) and prostatic volume (PV) were calculated using the ellipsoid formula. Statistical analysis was performed with the GraphPad Prism program (version 6.01). RESULTS: The fetuses presented PV between 6.1 and 297.18 mm2  (mean = 77.98 mm3 ). Linear regression analysis indicated that the PV in these fetuses increased significantly and positively with fetal age (r2 = .3120; p < .0001). We did not observe significant differences between the TV (right testis: 0.39-63.94 mm3 ; mean = 19.84 mm3 ; left testis: ​​​​​​0.52-55.37 mm3 , mean = 17.25 mm3 ). Linear regression analysis also indicated that the right and left TV (right: r2 = .6649; p < .0001 and left: r2 = .6792; p < .001) increased significantly and positively with fetal age. CONCLUSION: The prostatic growth was slower during the second gestational trimester, with significant correlations with fetal biometric parameters. The testicular growth was moderate and showed a significant correlation with fetal parameters during the studied period in human fetuses.


Subject(s)
Fetal Development , Prostate/embryology , Testis/embryology , Anthropometry , Female , Fetal Weight , Gestational Age , Humans , Male , Organ Size , Pregnancy , Pregnancy Trimester, Second
6.
Dev Biol ; 447(2): 157-169, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30659795

ABSTRACT

DNA methyltransferase 1 (DNMT1) is required for embryogenesis but roles in late forming organ systems including the prostate, which emerges from the urethral epithelium, have not been fully examined. We used a targeted genetic approach involving a Shhcre recombinase to demonstrate requirement of epithelial DNA methyltransferase-1 (Dnmt1) in mouse prostate morphogenesis. Dnmt1 mutant urethral cells exhibit DNA hypomethylation, DNA damage, p53 accumulation and undergo cell cycle arrest and apoptosis. Urethral epithelial cells are disorganized in Dnmt1 mutants, leading to impaired prostate growth and maturation and failed glandular development. We evaluated oriented cell division as a mechanism of bud elongation and widening by demonstrating that mitotic spindle axes typically form parallel or perpendicular to prostatic bud elongation axes. We then deployed a ShhcreERT allele to delete Dnmt1 from a subset of urethral epithelial cells, creating mosaic mutants with which to interrogate the requirement for cell division in specific prostatic bud epithelial populations. DNMT1- cell distribution within prostatic buds is not random as would be expected in a process where DNMT1 was not required. Instead, replication competent DNMT1 + cells primarily accumulate in prostatic bud margins and tips while replication impeded DNMT1- cells accumulate in prostatic bud cores. Together, these results highlight the role of DNMT1 in regulating epithelial bud formation by maintaining cell cycle progression and survival of rapidly dividing urethral epithelial cells, which can be extended to the study of other developing epithelial organs. In addition, our results show that prostatic buds consist of two epithelial cell populations with distinct molecular and functional characteristics that could potentially contribute to specialized lineages in the adult prostate.


Subject(s)
Cell Cycle/physiology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Epithelial Cells/enzymology , Organogenesis/physiology , Prostate/embryology , Urethra/embryology , Animals , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Epithelial Cells/cytology , Male , Mice , Mice, Transgenic , Prostate/cytology , Urethra/cytology
7.
Dev Biol ; 446(2): 180-192, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30594504

ABSTRACT

The evolutionarily conserved transcription factor, Tbx18, is expressed in a dynamic pattern throughout embryonic and early postnatal life and plays crucial roles in the development of multiple organ systems. Previous studies have indicated that this dynamic function is controlled by an expansive regulatory structure, extending far upstream and downstream of the gene. With the goal of identifying elements that interact with the Tbx18 promoter in developing prostate, we coupled chromatin conformation capture (4C) and ATAC-seq from embryonic day 18.5 (E18.5) mouse urogenital sinus (UGS), where Tbx18 is highly expressed. The data revealed dozens of active chromatin elements distributed throughout a 1.5 million base pair topologically associating domain (TAD). To identify cell types contributing to this chromatin signal, we used lineage tracing methods with a Tbx18 Cre "knock-in" allele; these data show clearly that Tbx18-expressing precursors differentiate into wide array of cell types in multiple tissue compartments, most of which have not been previously reported. We also used a 209 kb Cre-expressing Tbx18 transgene, to partition enhancers for specific precursor types into two rough spatial domains. Within this central 209 kb compartment, we identified ECR1, previously described to regulate Tbx18 expression in ureter, as an active regulator of UGS expression. Together these data define the diverse fates of Tbx18+ precursors in prostate-associated tissues for the first time, and identify a highly active TAD controlling the gene's essential function in this tissue.


Subject(s)
Gene Expression Regulation, Developmental , Prostate/metabolism , Regulatory Elements, Transcriptional/genetics , T-Box Domain Proteins/genetics , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , High-Throughput Nucleotide Sequencing/methods , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Prostate/cytology , Prostate/embryology , T-Box Domain Proteins/metabolism , Urogenital System/cytology , Urogenital System/embryology , Urogenital System/metabolism
8.
Development ; 144(8): 1382-1398, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28400434

ABSTRACT

Prostate organogenesis is a complex process that is primarily mediated by the presence of androgens and subsequent mesenchyme-epithelial interactions. The investigation of prostate development is partly driven by its potential relevance to prostate cancer, in particular the apparent re-awakening of key developmental programs that occur during tumorigenesis. However, our current knowledge of the mechanisms that drive prostate organogenesis is far from complete. Here, we provide a comprehensive overview of prostate development, focusing on recent findings regarding sexual dimorphism, bud induction, branching morphogenesis and cellular differentiation.


Subject(s)
Hormones/metabolism , Organogenesis , Prostate/cytology , Prostate/embryology , Animals , Cell Differentiation , Epithelium/embryology , Humans , Male , Prostate/anatomy & histology , Sex Characteristics
9.
Development ; 144(12): 2282-2293, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28506996

ABSTRACT

In humans and rodents, the prostate gland develops from the embryonic urogenital sinus (UGS). The androgen receptor (AR) is thought to control the expression of morphogenetic genes in inductive UGS mesenchyme, which promotes proliferation and cytodifferentiation of the prostatic epithelium. However, the nature of the AR-regulated morphogenetic genes and the mechanisms whereby AR controls prostate development are not understood. Glial cell line-derived neurotrophic factor (GDNF) binds GDNF family receptor α1 (GFRα1) and signals through activation of RET tyrosine kinase. Gene disruption studies in mice have revealed essential roles for GDNF signaling in development; however, its role in prostate development is unexplored. Here, we establish novel roles of GDNF signaling in mouse prostate development. Using an organ culture system for prostate development and Ret mutant mice, we demonstrate that RET-mediated GDNF signaling in UGS increases proliferation of mesenchyme cells and suppresses androgen-induced proliferation and differentiation of prostate epithelial cells, inhibiting prostate development. We also identify Ar as a GDNF-repressed gene and Gdnf and Gfrα1 as androgen-repressed genes in UGS, thus establishing reciprocal regulatory crosstalk between AR and GDNF signaling in prostate development.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Prostate/embryology , Prostate/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Actins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Dihydrotestosterone/pharmacology , Female , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Morphogenesis/genetics , Morphogenesis/physiology , Organ Culture Techniques , Pregnancy , Prostate/cytology , Proto-Oncogene Proteins c-ret/genetics , Receptor Cross-Talk , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Signal Transduction
10.
Differentiation ; 107: 1-10, 2019.
Article in English | MEDLINE | ID: mdl-30927641

ABSTRACT

Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence also indicates a regulatory role of Notch signaling in prostate development, differentiation, and growth. However, the collaborative regulatory mechanisms of androgen and Notch signaling during prostate development, growth, and regeneration are largely unknown. Hairy and Enhancer of Split 1 (Hes1) is a transcriptional regulator of Notch signaling pathways, and its expression is responsive to Notch signaling. Hes1-expressing cells have been shown to possess the regenerative capability to repopulate a variety of adult tissues. In this study, we developed new mouse models to directly assess the role of the androgen receptor in prostatic Hes1-expressing cells. Selective deletion of AR expression in embryonic Hes1-expressing cells impeded early prostate development both in vivo and in tissue xenograft experiments. Prepubescent deletion of AR expression in Hes1-expressing cells resulted in prostate glands containing abnormalities in cell morphology and gland architecture. A population of castration-resistant Hes1-expressing cells was revealed in the adult prostate, with the ability to repopulate prostate epithelium following androgen supplementation. Deletion of AR in Hes1-expressing cells diminishes their regenerative ability. These lines of evidence demonstrate a critical role for the AR in Notch-responsive cells during the course of prostate development, morphogenesis, and regeneration, and implicate a mechanism underlying interaction between the androgen and Notch signaling pathways in the mouse prostate.


Subject(s)
Prostate/physiology , Receptors, Notch/metabolism , Regeneration , Transcription Factor HES-1 , Androgens/metabolism , Animals , Male , Mice , Models, Animal , Prostate/embryology , Receptors, Androgen/metabolism , Signal Transduction , Transcription Factor HES-1/biosynthesis , Transcription Factor HES-1/genetics
11.
Environ Toxicol ; 35(1): 15-26, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31454150

ABSTRACT

The prostate is an accessory reproductive gland that is sensitive to the action of exogenous compounds known as endocrine disrupters that alter normal hormonal function. Finasteride is a widely used chemical that acts to inhibit the conversion of testosterone in its most active form, dihydrotestosterone. It is known that intrauterine exposure to finasteride causes changes in the male prostate even at low dosages; however, it is not known whether these dosages are capable of causing changes in the female prostate, which is present in a large number of mammalian species, including humans. In the present study, histochemistry, immunohistochemistry, immunofluorescence, serological dosages, and three-dimensional reconstruction techniques were employed to evaluate the effects of intrauterine exposure to a low dose of finasteride (100 µg.BW/d) on postnatal prostate development in male and female Mongolian gerbils. The results indicate that the gerbil female prostate also undergoes alterations following intrauterine exposure to finasteride, exhibiting a thickening of periductal smooth muscle and increased stromal proliferation. There are also intersex differences in the impact of exposure on the expression of the androgen receptor, which was increased in males, and of the estrogen-α receptor, which was decreased in the male prostate but unchanged in females. Altogether, this study indicates there are sex differences in the effects of finasteride exposure even at low dosages.


Subject(s)
Embryonic Development/drug effects , Endocrine Disruptors/toxicity , Finasteride/toxicity , Genitalia, Female/drug effects , Gerbillinae/embryology , Prenatal Exposure Delayed Effects/chemically induced , Prostate/drug effects , Animals , Dose-Response Relationship, Drug , Female , Genitalia, Female/embryology , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prostate/embryology , Receptors, Androgen/metabolism , Reproduction/drug effects , Testosterone/metabolism
12.
Histochem Cell Biol ; 152(1): 35-45, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30976911

ABSTRACT

Prostate autonomic and sensory axons control glandular growth, fluid secretion, and smooth muscle contraction and are remodeled during cancer and inflammation. Morphogenetic signaling pathways reawakened during disease progression may drive this axon remodeling. These pathways are linked to proliferative activities in prostate cancer and benign prostate hyperplasia. However, little is known about which developmental signaling pathways guide axon investment into prostate. The first step in defining these pathways is pinpointing when axon subtypes first appear in prostate. We accomplished this by immunohistochemically mapping three axon subtypes (noradrenergic, cholinergic, and peptidergic) during fetal, neonatal, and adult stages of mouse prostate development. We devised a method for peri-prostatic axon density quantification and tested whether innervation is uniform across the proximo-distal axis of dorsal and ventral adult mouse prostate. Many axons directly interact with or innervate neuroendocrine cells in other organs, so we examined whether sensory or autonomic axons innervate neuroendocrine cells in prostate. We first detected noradrenergic, cholinergic, and peptidergic axons in prostate at embryonic day (E) 14.5. Noradrenergic and cholinergic axon densities are uniform across the proximal-distal axis of adult mouse prostate while peptidergic axons are denser in the periurethral and proximal regions. Peptidergic and cholinergic axons are closely associated with prostate neuroendocrine cells whereas noradrenergic axons are not. These results provide a foundation for understanding mouse prostatic axon development and organization and, provide strategies for quantifying axons during progression of prostate disease.


Subject(s)
Axons/metabolism , Prostate/embryology , Prostate/innervation , Animals , Axons/pathology , Male , Mice , Mice, Inbred C57BL , Prostate/cytology , Prostate/pathology
13.
Toxicol Pathol ; 47(8): 1038-1042, 2019 12.
Article in English | MEDLINE | ID: mdl-31662055

ABSTRACT

The purpose of this symposium report is to summarize information from a session 3 oral presentation at the Society of Toxicologic Pathology Annual Symposium in Raleigh, North Carolina. Mice are genetically tractable and are likely to play an important role in elucidating environmental, genetic, and aging-related mechanisms of urinary dysfunction in men. We and others have made significant strides in developing quantitative methods for assessing mouse urinary function and our collaborators recently showed that aging male mice, like men, develop urinary dysfunction. Yet, it remains unclear how mouse prostate anatomy and histology relate to urinary function. The purpose of this report is to share foundational resources for evaluating mouse prostate histology and urinary physiology from our recent publication "Impact of Sex, Androgens, and Prostate Size on C57BL/6J Mouse Urinary Physiology: Functional Assessment." We will begin with a review of prostatic embryology in men and mice, then move to comparative histology resources, and conclude with quantitative measures of rodent urinary physiology.


Subject(s)
Androgens/metabolism , Organogenesis/physiology , Prostate/embryology , Urinary Bladder/physiology , Urinary Tract Physiological Phenomena , Aging/physiology , Animals , Congresses as Topic , Humans , Male , Mice , Mice, Inbred C57BL , Organ Size/physiology , Prostate/anatomy & histology , Prostate/metabolism , Species Specificity , Urinary Bladder/anatomy & histology , Urinary Bladder/metabolism
14.
Dev Dyn ; 247(5): 679-685, 2018 05.
Article in English | MEDLINE | ID: mdl-29368411

ABSTRACT

BACKGROUND: Prostate ductal branching morphogenesis involves a complex spatiotemporal regulation of cellular proliferation and remodeling of the extracellular matrix (ECM) around the developing ducts. Decorin (Dcn) is a small leucine-rich proteoglycan known to sequester several growth factors and to act as a tumor suppressor in prostate cancer. RESULTS: Dcn expression in the developing prostate paralleled branching morphogenesis and was dynamically regulated by androgen and Hedgehog (Hh) signaling. DCN colocalized with collagen in the periductal stroma and acellular interstitium. Exogenous DCN decreased epithelial proliferation in ex vivo organ cultures of developing prostate, whereas genetic ablation of Dcn resulted in increased epithelial proliferation in the developing prostate. CONCLUSIONS: Dcn expression and localization in the developing prostate is consistent with a primary role in organizing collagen around the developing ducts. Regulation of Dcn expression appears to be complex, involving both androgen and Hh signaling. The growth inhibitory effect of Dcn suggests a unique linkage between a structural proteoglycan and epithelial growth regulation. This may serve to coordinate two elements of the morphogenetic process: ductal growth and organization of the collagen matrix around the nascent duct. Developmental Dynamics 247:679-685, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Decorin/metabolism , Prostate/metabolism , Signal Transduction/physiology , Animals , Decorin/genetics , Female , Male , Mice , Morphogenesis/physiology , Organ Culture Techniques , Organogenesis/physiology , Prostate/embryology , Prostate/growth & development
15.
J Biol Chem ; 292(5): 2021-2031, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28003366

ABSTRACT

The histogenesis of prostatic neuroendocrine cells is controversial: a stem cell hypothesis with a urogenital sinus-derived progeny of all prostatic epithelial cells is opposed by a dual origin hypothesis, favoring the derivation of neuroendocrine cells from the neural crest, with the secretory and basal cells being of urogenital sinus origin. A computer-assisted 3D reconstruction was used to analyze the distribution of chromogranin A immunoreactive cells in serial sections of human fetal prostate specimens (gestation weeks 18 and 25). Immunohistochemical double labeling studies with YFP and serotonin antisera combined with electron microscopy were carried out on double-transgenic Wnt1-Cre/ROSA26-YFP mice showing stable YFP expression in all neural crest-derived cell populations despite loss of Wnt1 expression. 3D reconstruction of the distribution pattern of neuroendocrine cells in the human fetal prostate indicates a migration of paraganglionic cells passing the stroma and reaching the prostate ducts. Double-transgenic mice showed 55% double labeling of periurethral neuroendocrine cells expressing both serotonin and YFP, whereas single serotonin labeling was observed in 36% and exclusive YFP labeling in 9%. The results favor the assumption of a major fraction of neural crest-derived neuroendocrine cells in both the human and murine prostates.


Subject(s)
Neural Crest/embryology , Neuroendocrine Cells/metabolism , Prostate/embryology , Animals , Humans , Male , Mice , Mice, Transgenic , Neural Crest/cytology , Neuroendocrine Cells/cytology , Prostate/cytology , Wnt1 Protein/biosynthesis , Wnt1 Protein/genetics
16.
Cell Biol Int ; 41(11): 1184-1193, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28741838

ABSTRACT

The effects of intrauterine exposure to 17ß-oestradiol (E2) are well studied for the male prostate and there are accumulating evidences that the exposure to high dosages leads to a hypomorphic development. However, there is a lack of information about the effects of intrauterine exposure to E2 in the prostate of rodent females, and such research becomes relevant in view of the presence of functional prostate in a proportion of women, and the morphophysiological similarities between the prostate of female rodents and the prostate of women. This study uses histochemical, immunohistochemical, immunofluorescence and three-dimensional (3D) reconstruction techniques to evaluate the effects of intrauterine exposure to E2 (500 BW/d) on neonatal prostate development in both male and female gerbils. It was verified that intrauterine exposure to E2 promotes epithelial proliferation and growth of prostatic budding in females, whereas in males the prostatic budding shows hypomorphic growth in the VMP (Ventral Mesenchymal Pad) as well as reduced epithelial proliferation. Together, the data demonstrate that intrauterine exposure to E2 causes different effects on male and female prostates of the gerbil even at the early postnatal development of the gland.


Subject(s)
Estradiol/metabolism , Estradiol/pharmacology , Prostate/drug effects , Animals , Animals, Newborn/embryology , Animals, Newborn/metabolism , Endocrine Disruptors/metabolism , Endocrine Disruptors/pharmacology , Female , Gerbillinae/embryology , Male , Pregnancy , Prenatal Exposure Delayed Effects , Prostate/embryology , Receptors, Androgen/drug effects , Receptors, Estrogen/drug effects , Sex Factors
17.
Dev Biol ; 400(1): 94-104, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25641695

ABSTRACT

The prostate gland plays an important role in male reproduction, and is also an organ prone to diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. The prostate consists of ducts with an inner layer of epithelium surrounded by stroma. Reciprocal signaling between these two cell compartments is instrumental to normal prostatic development, homeostasis, regeneration, as well as tumor formation. Hedgehog (HH) signaling is a master regulator in numerous developmental processes. In many organs, HH plays a key role in epithelial-mesenchymal signaling that regulates organ growth and tissue differentiation, and abnormal HH signaling has been implicated in the progression of various epithelial carcinomas. In this review, we focus on recent studies exploring the multipotency of endogenous postnatal and adult epithelial and stromal stem cells and studies addressing the role of HH in prostate development and cancer. We discuss the implications of the results for a new understanding of prostate development and disease. Insight into the cellular and molecular mechanisms underlying epithelial-mesenchymal growth regulation should provide a basis for devising innovative therapies to combat diseases of the prostate.


Subject(s)
Adult Stem Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Prostate/embryology , Prostate/growth & development , Prostatic Neoplasms/physiopathology , Signal Transduction/physiology , Animals , Humans , Male , Mice , Prostate/metabolism , Prostatic Neoplasms/metabolism , Species Specificity
18.
Int J Exp Pathol ; 97(4): 310-316, 2016 08.
Article in English | MEDLINE | ID: mdl-27469444

ABSTRACT

Developmental toxicity caused by environmental exposure to heavy metals during the perinatal period has raised questions about offspring health. Cadmium (Cd) is an endocrine-disrupting chemical with the potential to interfere with morphogenesis and susceptibility to diseases in reproductive organs. Taking into account that in the rat prostate morphogenesis occurs during the perinatal period, and that pregnant females absorb and retain more dietary Cd than their non-pregnant counterparts, it is important to understand the effects of perinatal Cd exposure on the adult rat prostate. Therefore this study investigated the effects of gestational and lactational Cd exposure on adult offspring rat prostate histopathology. Pregnant rats (n = 20) were divided into two groups: Control (treated with aqueous solution of sodium acetate 10 mg/l) and treated (treated with aqueous solution of cadmium acetate 10 mg/l) administered in the drinking water. After weaning, male offspring from different litters (n = 10) received food and water 'ad libitum'. The animals were euthanized at postnatal day 90 (PND90), the ventral prostates (VPs) were removed, weighed and examined histopathologically. Blood was collected for the measurement of testosterone (T) levels. Immunohistochemistry for androgen receptor (AR) and Ki67, and a TUNEL assay were performed. There were no differences in T levels, cell proliferation and apoptosis indexes, or AR immunostaining between the experimental groups. Stromal inflammatory foci and multifocal inflammation increased significantly in the treated group. These changes were associated with inflammatory reactive epithelial atypia and stromal fibrillar rearrangement. In conclusion, VP was permanently affected by perinatal Cd exposition, with increased incidence of inflammatory disorders with ageing.


Subject(s)
Acetates/toxicity , Cadmium/toxicity , Endocrine Disruptors/toxicity , Prenatal Exposure Delayed Effects , Prostate/drug effects , Animals , Female , Lactation , Male , Maternal-Fetal Exchange , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prostate/embryology , Prostate/metabolism , Prostate/pathology , Rats, Wistar , Receptors, Androgen/metabolism , Testosterone/blood
19.
Environ Toxicol ; 31(12): 1740-1750, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26443714

ABSTRACT

Substances that mimic endogenous hormones may alter the cell signaling that govern prostate development and predispose it to developing lesions in adult and senile life. Bisphenol A is able to mimic estrogens, and studies have demonstrated that low levels of exposure to this compound have caused alterations during prostate development. The aim of this study was to describe the prostate development in both male and female neonatal gerbils in normal conditions and under exposure to BPA during intrauterine life, and also to analyze whether the effects of intrauterine exposure to BPA remain in adulthood. Morphological, stereological, three-dimensional reconstruction, and immunohistochemical methods were employed. The results demonstrated that in 1-day-old normal gerbils, the female paraurethral glands and the male ventral lobe are morphologically similar, although its tissue components-epithelial buds (EB), periurethral mesenchyme (PeM), paraurethral mesenchyme (PaM) or ventral mesenchymal pad (VMP), and smooth muscle (SM)-have presented different immunolabeling pattern for androgen receptor (AR), and for proliferating cell nuclear antigen (PCNA). Moreover, we observed a differential response of male and female prostate to intrauterine BPA exposure. In 1-day-old males, the intrauterine exposure to BPA caused a decrease of AR-positive cells in the PeM and SM, and a decrease of the proliferative status in the EB. In contrast, no morphological alterations were observed in ventral prostate of adult males. In 1-day-old females, BPA exposure promoted an increase of estrogen receptor alpha (ERα) positive cells in PeM and PaM, a decrease of AR-positive cells in EB and PeM, besides a reduction of cell proliferation in EB. Additionally, the adult female prostate of BPA-exposed animals presented an increase of AR- and PCNA-positive cells. These results suggest that the prostate of female gerbils were more susceptible to the intrauterine BPA effects, since they became more proliferative in adult life. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1740-1750, 2016.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Phenols/toxicity , Urogenital System/drug effects , Age Factors , Animals , Animals, Newborn , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Estrogen Receptor alpha/metabolism , Female , Gerbillinae , Male , Maternal Exposure/adverse effects , Mesoderm/drug effects , Mesoderm/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Prostate/cytology , Prostate/drug effects , Prostate/embryology , Prostate/growth & development , Receptors, Androgen/metabolism , Sex Factors , Urogenital System/cytology , Urogenital System/embryology , Urogenital System/growth & development
20.
Dev Biol ; 395(2): 209-17, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25261715

ABSTRACT

The mammalian urogenital sinus (UGS) develops in a sex specific manner, giving rise to the prostate in the male and the sinus vagina in the embryonic female. Androgens, produced by the embryonic testis, have been shown to be crucial to this process. In this study we show that retinoic acid signaling is required for the initial stages of bud development from the male UGS. Enzymes involved in retinoic acid synthesis are expressed in the UGS mesenchyme in a sex specific manner and addition of ligand to female tissue is able to induce prostate-like bud formation in the absence of androgens, albeit at reduced potency. Functional studies in mouse organ cultures that faithfully reproduce the initiation of prostate development indicate that one of the roles of retinoic acid signaling in the male is to inhibit the expression of Inhba, which encodes the ßA subunit of Activin, in the UGS mesenchyme. Through in vivo genetic analysis and culture studies we show that inhibition of Activin signaling in the female UGS leads to a similar phenotype to that of retinoic acid treatment, namely bud formation in the absence of androgens. Our data also reveals that both androgens and retinoic acid have extra independent roles to that of repressing Activin signaling in the development of the prostate during fetal stages. This study identifies a novel role for retinoic acid as a mesenchymal factor that acts together with androgens to determine the position and initiation of bud development in the male UGS epithelia.


Subject(s)
Activins/metabolism , Organogenesis/physiology , Signal Transduction/physiology , Tretinoin/metabolism , Urogenital System/embryology , Activins/antagonists & inhibitors , Animals , DNA Primers , Female , Immunohistochemistry , In Situ Hybridization , Inhibin-beta Subunits/antagonists & inhibitors , Male , Mice , Prostate/embryology , Real-Time Polymerase Chain Reaction , Sex Factors , Tretinoin/pharmacology , Urogenital System/metabolism , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL