Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 379
Filtrar
1.
Dev Biol ; 473: 50-58, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33529704

RESUMO

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.


Assuntos
Próstata/metabolismo , Próstata/patologia , Células-Tronco/metabolismo , Antagonistas de Androgênios/metabolismo , Androgênios/metabolismo , Animais , Diferenciação Celular , Células Epiteliais/metabolismo , Humanos , Masculino , Organogênese , Próstata/embriologia , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo
2.
Prostate ; 81(3): 214-219, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33393689

RESUMO

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.


Assuntos
Desenvolvimento Fetal , Próstata/embriologia , Testículo/embriologia , Antropometria , Feminino , Peso Fetal , Idade Gestacional , Humanos , Masculino , Tamanho do Órgão , Gravidez , Segundo Trimestre da Gravidez
3.
Dev Dyn ; 250(5): 618-628, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33325097

RESUMO

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.


Assuntos
Heparitina Sulfato/biossíntese , Próstata/metabolismo , Animais , Glucuronidase/metabolismo , Humanos , Masculino , Próstata/embriologia , Transdução de Sinais
4.
Environ Toxicol ; 35(1): 15-26, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31454150

RESUMO

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.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Finasterida/toxicidade , Genitália Feminina/efeitos dos fármacos , Gerbillinae/embriologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Próstata/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Genitália Feminina/embriologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Próstata/embriologia , Receptores Androgênicos/metabolismo , Reprodução/efeitos dos fármacos , Testosterona/metabolismo
5.
J Pediatr Surg ; 55(10): 2221-2225, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31708209

RESUMO

BACKGROUND: To compare the growth of the prostate in anencephalic, prune belly syndrome (PBS) and control fetuses. METHODS: We studied 35 prostates from normal human fetuses aged 11-22 weeks postconception (WPC); 15 from anencephalic fetuses aged 13-19 WPC; and 6 from PBS fetuses aged 13-31WPC. After prostate dissection, we evaluated the prostate length, width and thickness with the aid of a computer program (Image Pro and Image J). The fetal prostate volume (PV) was calculated using the ellipsoid formula: PV = [length × thickness × width] × 0.523. The prostates were dissected and the PV was measured with the aid of the same computer program. Means were statistically compared using the unpaired t-test and linear regression was performed. RESULTS: In 2 PBS fetuses we observed prostatic atresia. We did not observe significant differences in PV when comparing the control group (PV: 6.1 to 313.81 mm, mean = 70.85 mm: SD = 71.43 mm) with anencephalic fetuses: p = 0.3575 (PV: 5.1 to 159.11 mm, mean = 42.94 mm; SD = 40.11 mm) and PBS fetuses: p > 0.999 (PV: 10.89 to 148.71 mm, mean = 55.4 mm; SD = 63.64 mm). The linear regression analysis indicated that the PV in the control group (r2 = 0.3096; p = 0.0004), anencephalic group (r2 = 0.3778; p = 0.0148) and PBS group (r2 = 0.9821; p < 0.009) increased significantly and positively with fetal age (p < 0.0001). CONCLUSIONS: We did not observe significant differences in development of the prostate in fetuses with anencephaly and in 2/3 of fetuses with PBS during the fetal period studied. In 1/3 of the PBS fetuses, the prostate had important atresia. LEVEL OF EVIDENCE: Level III.


Assuntos
Anencefalia/embriologia , Próstata/embriologia , Próstata/crescimento & desenvolvimento , Síndrome do Abdome em Ameixa Seca/embriologia , Feto/embriologia , Idade Gestacional , Humanos , Masculino , Tamanho do Órgão
6.
Toxicol Pathol ; 47(8): 1038-1042, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31662055

RESUMO

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.


Assuntos
Androgênios/metabolismo , Organogênese/fisiologia , Próstata/embriologia , Bexiga Urinária/fisiologia , Fenômenos Fisiológicos do Sistema Urinário , Envelhecimento/fisiologia , Animais , Congressos como Assunto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/fisiologia , Próstata/anatomia & histologia , Próstata/metabolismo , Especificidade da Espécie , Bexiga Urinária/anatomia & histologia , Bexiga Urinária/metabolismo
7.
Development ; 146(20)2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31575645

RESUMO

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.


Assuntos
Células-Tronco Multipotentes/citologia , Próstata/citologia , Próstata/embriologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células/fisiologia , Células Cultivadas , Masculino , Camundongos , Células-Tronco Multipotentes/metabolismo , Organogênese/fisiologia , Próstata/metabolismo
8.
Histochem Cell Biol ; 152(1): 35-45, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30976911

RESUMO

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.


Assuntos
Axônios/metabolismo , Próstata/embriologia , Próstata/inervação , Animais , Axônios/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Próstata/citologia , Próstata/patologia
9.
Differentiation ; 107: 1-10, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30927641

RESUMO

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.


Assuntos
Próstata/fisiologia , Receptores Notch/metabolismo , Regeneração , Fatores de Transcrição HES-1 , Androgênios/metabolismo , Animais , Masculino , Camundongos , Modelos Animais , Próstata/embriologia , Receptores Androgênicos/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/biossíntese , Fatores de Transcrição HES-1/genética
10.
Stem Cell Res ; 35: 101405, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30776675

RESUMO

Human induced pluripotent stem cell line was generated from commercially available primary human prostate fibroblasts HPrF derived from a fetus, aged 18-24 weeks of gestation. The fibroblast cell line was reprogrammed with Yamanaka factors (OCT4, SOX2, c-MYC, KLF4) using CytoTune™-iPS 2.0 Sendai Reprogramming Kit. Pluripotency of the derived transgene-free iPS cell line was confirmed both in vitro by detecting the expression of factors of pluripotency on a single-cell level, and in vivo using teratoma formation assay. This iPS cell line will be a useful tool for studying both normal prostate development and prostate cancer disease.


Assuntos
Técnicas de Reprogramação Celular , Feto , Fibroblastos , Células-Tronco Pluripotentes Induzidas , Próstata , Reprogramação Celular , Feto/citologia , Feto/embriologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Masculino , Próstata/citologia , Próstata/embriologia
11.
Dev Biol ; 447(2): 157-169, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30659795

RESUMO

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.


Assuntos
Ciclo Celular/fisiologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Células Epiteliais/enzimologia , Organogênese/fisiologia , Próstata/embriologia , Uretra/embriologia , Animais , Sobrevivência Celular , DNA (Citosina-5-)-Metiltransferase 1/genética , Células Epiteliais/citologia , Masculino , Camundongos , Camundongos Transgênicos , Próstata/citologia , Uretra/citologia
12.
Dev Biol ; 446(2): 180-192, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30594504

RESUMO

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.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Próstata/metabolismo , Elementos Reguladores de Transcrição/genética , Proteínas com Domínio T/genética , Animais , Sítios de Ligação/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Cromatina/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Masculino , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Próstata/citologia , Próstata/embriologia , Proteínas com Domínio T/metabolismo , Sistema Urogenital/citologia , Sistema Urogenital/embriologia , Sistema Urogenital/metabolismo
13.
Nat Rev Urol ; 15(11): 703-715, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30214054

RESUMO

The prostate is the only organ in a man that continues to grow with age. John McNeal proposed, 40 years ago, that this BPH is characterized by an age-related reinitiation of benign neoplastic growth selectively in developmentally abortive distal ducts within the prostate transition-periurethral zone (TPZ), owing to a reawakening of inductive stroma selectively within these zones. An innovative variant of this hypothesis is that, owing to its location, the TPZ is continuously exposed to urinary components and/or autoantigens, which produces an inflammatory TPZ microenvironment that promotes recruitment of bone marrow-derived mesenchymal stem cells (MSCs) and generates a paracrine-inductive stroma that reinitiates benign neoplastic nodular growth. In support of this hypothesis, MSCs infiltrate human BPH tissue and have the ability to stimulate epithelial stem cell growth. These results provide a framework for defining both the aetiology of BPH in ageing men and insights into new therapeutic approaches.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Próstata/embriologia , Hiperplasia Prostática/embriologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Biomarcadores/metabolismo , Humanos , Masculino , Próstata/metabolismo , Próstata/patologia , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Hiperplasia Prostática/terapia , Transdução de Sinais
14.
Dev Dyn ; 247(5): 679-685, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29368411

RESUMO

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.


Assuntos
Decorina/metabolismo , Próstata/metabolismo , Transdução de Sinais/fisiologia , Animais , Decorina/genética , Feminino , Masculino , Camundongos , Morfogênese/fisiologia , Técnicas de Cultura de Órgãos , Organogênese/fisiologia , Próstata/embriologia , Próstata/crescimento & desenvolvimento
15.
Birth Defects Res ; 110(3): 190-227, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29063715

RESUMO

This review describes pre- and postnatal development of the male reproductive system in humans and laboratory animals, and highlights species differences in the timing and control of hormonal and morphologic events. Major differences are that the fetal testis is dependent on gonadotropins in humans, but is independent of such in rats; humans have an extended postnatal quiescent period, whereas rats exhibit no quiescence; and events such as secretion by the prostate and seminal vesicles, testicular descent, and the appearance of spermatogonia are all prenatal events in humans, but are postnatal events in rats. Major differences in the timing of the developmental sequence between rats and humans include: gonocyte transformation period (rat: postnatal day 0-9; human: includes gestational week 22 to 9 months of age); masculinization programming window (rat: gestational day 15.5-17.5; human: gestational week 9-14); and mini-puberty (rat: 0-6 hr after birth; human: 3-6 months of age). Endocrine disruptors can cause unique lesions in the prenatal and early postnatal testis; therefore, it is important to consider the differences in the timing of the developmental sequence when designing preclinical studies as identification of windows of sensitivity for endocrine disruption or toxicants will aid in interpretation of results and provide clues to a mode of action. Birth Defects Research 110:190-227, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Próstata/embriologia , Próstata/crescimento & desenvolvimento , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Animais , Disruptores Endócrinos/toxicidade , Humanos , Masculino , Ratos , Especificidade da Espécie
16.
Artigo em Inglês | MEDLINE | ID: mdl-28289061

RESUMO

Epithelial cells contribute to the development of various vital organs by generating tubular and/or glandular architectures. The fully developed forms of ductal organs depend on processes of branching morphogenesis, whereby frequency, total number, and complexity of the branching tissue define the final architecture in the organ. Some ductal tissues, like the mammary gland during pregnancy and lactation, disintegrate and regenerate through periodic cycles. Differentiation of branched epithelia is driven by antagonistic actions of parallel growth factor systems that mediate epithelial-mesenchymal communication. Transforming growth factor-ß (TGF-ß) family members and their extracellular antagonists are prominently involved in both normal and disease-associated (e.g., malignant or fibrotic) ductal tissue patterning. Here, we discuss collective knowledge that permeates the roles of TGF-ß family members in the control of the ductal tissues in the vertebrate body.


Assuntos
Morfogênese , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Transição Epitelial-Mesenquimal , Feminino , Humanos , Pulmão/embriologia , Masculino , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Humanas/embriologia , Organogênese , Pâncreas/embriologia , Próstata/embriologia , Glândulas Salivares/embriologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-29229667

RESUMO

The prostate is a male exocrine gland that secretes components of the seminal fluid. In men, prostate tumors are one of the most prevalent cancers. Studies on the development of the prostate have given a better understanding of the processes and genes that are important in the formation of this organ and have provided insights into the mechanisms of prostate tumorigenesis. These developmental studies have provided evidence that some of the genes and signaling pathways involved in development are reactivated or deregulated during prostate cancer. The prostate goes through a number of different stages during organogenesis, which include organ specification, epithelial budding, branching morphogenesis, canalization, and cytodifferentiation. During development, these processes are tightly regulated, many of which are controlled by the male hormone androgens. The majority of prostate tumors remain hormone regulated, and antiandrogen therapy is a first-line therapy, highlighting the important link between prostate organogenesis and cancer. In this review, we describe some of the data on genes that have important roles during prostate development that also have strong evidence linking them to prostate cancer.


Assuntos
Organogênese/fisiologia , Próstata/embriologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Masculino , Camundongos , Modelos Animais , Próstata/citologia , Próstata/crescimento & desenvolvimento , Neoplasias da Próstata/etiologia , Receptores Androgênicos/fisiologia , Fatores de Transcrição SOX9/fisiologia , Transdução de Sinais/fisiologia , Testosterona/fisiologia
18.
Cell ; 171(1): 242-255.e27, 2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28938116

RESUMO

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.


Assuntos
Rim/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Modelos Biológicos , Morfogênese , Próstata/crescimento & desenvolvimento , Animais , Feminino , Humanos , Rim/embriologia , Masculino , Glândulas Mamárias Humanas/embriologia , Camundongos , Próstata/embriologia
19.
Cell Biol Int ; 41(11): 1184-1193, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28741838

RESUMO

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.


Assuntos
Estradiol/metabolismo , Estradiol/farmacologia , Próstata/efeitos dos fármacos , Animais , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/metabolismo , Disruptores Endócrinos/metabolismo , Disruptores Endócrinos/farmacologia , Feminino , Gerbillinae/embriologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Próstata/embriologia , Receptores Androgênicos/efeitos dos fármacos , Receptores de Estrogênio/efeitos dos fármacos , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...