Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Toxicol Sci ; 199(2): 210-226, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38526210

RESUMO

In avian embryos, xenoestrogens induce abnormalities in reproductive organs, particularly the testes and Müllerian ducts (MDs). However, the molecular mechanisms remain poorly understood. We investigated the effects of ethynylestradiol (EE2) exposure on gene expression associated with reproductive organ development in Japanese quail embryos. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis revealed that the left testis containing ovary-like tissues following EE2 exposure highly expressed the genes for steroidogenic enzymes (P450scc, P45017α, lyase, and 3ß-HSD) and estrogen receptor-ß, compared to the right testis. No asymmetry was found in these gene expression without EE2. EE2 induced hypertrophy in female MDs and suppressed atrophy in male MDs on both sides. RNA sequencing analysis of female MDs showed 1,366 differentially expressed genes between developing left MD and atrophied right MD in the absence of EE2, and these genes were enriched in Gene Ontology terms related to organogenesis, including cell proliferation, migration and differentiation, and angiogenesis. However, EE2 reduced asymmetrically expressed genes to 21. RT-qPCR analysis indicated that genes promoting cell cycle progression and oncogenesis were more highly expressed in the left MD than in the right MD, but EE2 eliminated such asymmetric gene expression by increasing levels on the right side. EE2-exposed males showed overexpression of these genes in both MDs. This study reveals part of the molecular basis of xenoestrogen-induced abnormalities in avian reproductive organs, where EE2 may partly feminize gene expression in the left testis, developing as the ovotestis, and induce bilateral MD malformation by canceling asymmetric gene expression underlying MD development.


Assuntos
Coturnix , Etinilestradiol , Regulação da Expressão Gênica no Desenvolvimento , Ductos Paramesonéfricos , Testículo , Animais , Masculino , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/embriologia , Testículo/patologia , Coturnix/embriologia , Coturnix/genética , Etinilestradiol/toxicidade , Ductos Paramesonéfricos/efeitos dos fármacos , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/anormalidades , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Feminização/induzido quimicamente , Feminização/genética
2.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-33530552

RESUMO

The reproductive tract in mammals emerges from two ductal systems during embryogenesis: Wolffian ducts (WDs) and Mullerian ducts (MDs). Most of the female reproductive tract (FRT) including the oviducts, uterine horn and cervix, originate from MDs. It is widely accepted that the formation of MDs depends on the preformed WDs within the urogenital primordia. Here, we found that the WD mesenchyme under the regulation of Hedgehog (Hh) signaling is closely related to the developmental processes of the FRT during embryonic and postnatal periods. Deficiency of Sonic hedgehog (Shh), the only Hh ligand expressed exclusively in WDs, prevents the MD mesenchyme from affecting uterine growth along the radial axis. The in vivo cell tracking approach revealed that after WD regression, distinct cells responding to WD-derived Hh signal continue to exist in the developing FRT and gradually contribute to the formation of various tissues such as smooth muscle, endometrial stroma and vascular vessel, in the mouse uterus. Our study thus provides a novel developmental mechanism of FRT relying on WD.


Assuntos
Genitália Feminina/embriologia , Genitália Feminina/metabolismo , Proteínas Hedgehog/metabolismo , Organogênese , Transdução de Sinais , Útero/embriologia , Útero/metabolismo , Animais , Biomarcadores , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Modelos Biológicos , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Organogênese/genética
3.
BMC Genomics ; 21(1): 688, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33008304

RESUMO

BACKGROUND: Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract in vertebrates. Many disorders of female reproduction can be attributed to anomalies of Müllerian duct development. However, the molecular genetics of Müllerian duct formation is poorly understood and most disorders of duct development have unknown etiology. In this study, we describe for the first time the transcriptional landscape of the embryonic Müllerian duct, using the chicken embryo as a model system. RNA sequencing was conducted at 1 day intervals during duct formation to identify developmentally-regulated genes, validated by in situ hybridization. RESULTS: This analysis detected hundreds of genes specifically up-regulated during duct morphogenesis. Gene ontology and pathway analysis revealed enrichment for developmental pathways associated with cell adhesion, cell migration and proliferation, ERK and WNT signaling, and, interestingly, axonal guidance. The latter included factors linked to neuronal cell migration or axonal outgrowth, such as Ephrin B2, netrin receptor, SLIT1 and class A semaphorins. A number of transcriptional modules were identified that centred around key hub genes specifying matrix-associated signaling factors; SPOCK1, HTRA3 and ADGRD1. Several novel regulators of the WNT and TFG-ß signaling pathway were identified in Müllerian ducts, including APCDD1 and DKK1, BMP3 and TGFBI. A number of novel transcription factors were also identified, including OSR1, FOXE1, PRICKLE1, TSHZ3 and SMARCA2. In addition, over 100 long non-coding RNAs (lncRNAs) were expressed during duct formation. CONCLUSIONS: This study provides a rich resource of new candidate genes for Müllerian duct development and its disorders. It also sheds light on the molecular pathways engaged during tubulogenesis, a fundamental process in embryonic development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Ductos Paramesonéfricos/metabolismo , Transcriptoma , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Embrião de Galinha , Feminino , Ductos Paramesonéfricos/embriologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Toxicol Environ Health A ; 83(2): 66-81, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-32077375

RESUMO

Bisphenol A (BPA) and phthalate diesters are ubiquitous environmental contaminants. While these compounds have been reported as reproductive toxicants, their effects may partially be attributed to metabolites. The aim of this study was to examine reproductive organ development in chicken embryos exposed to the BPA metabolite, 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP; 100 µg/g egg) or a human-relevant mixture of 4 phthalate monoesters (85 µg/g egg). The mixture was designed within the EU project EDC-MixRisk based upon a negative association with anogenital distance in boys at 21 months of age in a Swedish pregnancy cohort. Chicken embryos were exposed in ovo from an initial stage of gonad differentiation (embryonic day 4) and dissected two days prior to anticipated hatching (embryonic day 19). No discernible effects were noted on reproductive organs in embryos exposed to the mixture. MBP-treated males exhibited retention of Müllerian ducts and feminization of the left testicle, while MBP-administered females displayed a diminished the left ovary. In the left testicle of MBP-treated males, mRNA expression of female-associated genes was upregulated while the testicular marker gene SOX9 was downregulated, corroborating a feminizing effect by MBP. Our results demonstrate that MBP, but not the phthalate monoester mixture, disrupts both male and female reproductive organ development in an avian embryo model.


Assuntos
Compostos Benzidrílicos/metabolismo , Compostos Benzidrílicos/toxicidade , Fenóis/metabolismo , Fenóis/toxicidade , Ácidos Ftálicos/química , Processos de Determinação Sexual/efeitos dos fármacos , Animais , Compostos Benzidrílicos/química , Embrião de Galinha , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Masculino , Ductos Paramesonéfricos/efeitos dos fármacos , Ductos Paramesonéfricos/embriologia , Ovário/efeitos dos fármacos , Ovário/embriologia , Fenóis/química , Ácidos Ftálicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Testículo/efeitos dos fármacos , Testículo/embriologia
5.
J Endocrinol ; 244(2): 395-413, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31829965

RESUMO

The embryonic Müllerian ducts give rise to the female reproductive tract (fallopian tubes, uterus and upper vagina in humans, the oviducts in birds). Embryonic Müllerian ducts initially develop in both sexes, but later regress in males under the influence of anti-Müllerian hormone. While the molecular and endocrine control of duct regression in males have been well studied, early development of the ducts in both sexes is less well understood. Here, we describe a novel role for the adhesion G protein-coupled receptor, GPR56, in development of the Müllerian ducts in the chicken embryo. GPR56 is expressed in the ducts of both sexes from early stages. The mRNA is present during the elongation phase of duct formation, and it is restricted to the inner Müllerian duct epithelium. The putative ligand, Collagen III, is abundantly expressed in the Müllerian duct at the same developmental stages. Knockdown of GPR56 expression using in ovo electroporation results in variably truncated ducts, with a loss of expression of both epithelial and mesenchymal markers of duct development. Over-expression of GPR56 in vitro results in enhanced cell proliferation and cell migration. These results show that GPR56 plays an essential role in avian Müllerian duct development through the regulation of duct elongation.


Assuntos
Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Proteínas Aviárias/genética , Proliferação de Células , Embrião de Galinha , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Receptores Acoplados a Proteínas G/genética
6.
Elife ; 82019 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-31232694

RESUMO

The Mullerian ducts are the anlagen of the female reproductive tract, which regress in the male fetus in response to MIS. This process is driven by subluminal mesenchymal cells expressing Misr2, which trigger the regression of the adjacent Mullerian ductal epithelium. In females, these Misr2+ cells are retained, yet their contribution to the development of the uterus remains unknown. Here, we report that subluminal Misr2+ cells persist postnatally in the uterus of rodents, but recede by week 37 of gestation in humans. Using single-cell RNA sequencing, we demonstrate that ectopic postnatal MIS administration inhibits these cells and prevents the formation of endometrial stroma in rodents, suggesting a progenitor function. Exposure to MIS during the first six days of life, by inhibiting specification of the stroma, dysregulates paracrine signals necessary for uterine development, eventually resulting in apoptosis of the Misr2+ cells, uterine hypoplasia, and complete infertility in the adult female.


Assuntos
Hormônio Antimülleriano/metabolismo , Ductos Paramesonéfricos/embriologia , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Útero/embriologia , Animais , Sequência de Bases , Feminino , Fertilidade , Perfilação da Expressão Gênica , Camundongos Endogâmicos C57BL
7.
Genetics ; 211(1): 219-233, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30446521

RESUMO

In laboratory strains of zebrafish, sex determination occurs in the absence of a typical sex chromosome and it is not known what regulates the proportion of animals that develop as males or females. Many sex determination and gonad differentiation genes that act downstream of a sex chromosome are well conserved among vertebrates, but studies that test their contribution to this process have mostly been limited to mammalian models. In mammals, WNT4 is a signaling ligand that is essential for ovary and Müllerian duct development, where it antagonizes the male-promoting FGF9 signal. Wnt4 is well conserved across all vertebrates, but it is not known if Wnt4 plays a role in sex determination and/or the differentiation of sex organs in nonmammalian vertebrates. This question is especially interesting in teleosts, such as zebrafish, because they lack an Fgf9 ortholog. Here we show that wnt4a is the ortholog of mammalian Wnt4, and that wnt4b was present in the last common ancestor of humans and zebrafish, but was lost in mammals. We show that wnt4a loss-of-function mutants develop predominantly as males and conclude that wnt4a activity promotes female sex determination and/or differentiation in zebrafish. Additionally, both male and female wnt4a mutants are sterile due to defects in reproductive duct development. Together these results strongly argue that Wnt4a is a conserved regulator of female sex determination and reproductive duct development in mammalian and nonmammalian vertebrates.


Assuntos
Ductos Paramesonéfricos/metabolismo , Diferenciação Sexual , Proteína Wnt4/genética , Proteínas de Peixe-Zebra/genética , Animais , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Masculino , Ductos Paramesonéfricos/embriologia , Proteína Wnt4/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
8.
G Chir ; 39(4): 245-247, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30039793

RESUMO

AIM: According to the so far published literature, only one case of endometrial cancer in a patient with unicornuate uterus has been reported. This is a case report study, presenting a rare case of complex atypical endometrial hyperplasia in a woman with unicornuate uterus and multiple genitourinary anomalies. CASE REPORT: A 43-year old G1P1 woman presented with episodes of menometrorrhagia and anemia. She had previous surgical history of laparoscopy due to infertility, in which she was diagnosed with unicornuate uterus with a rudimentary left uterine horn and ipsilateral ectopic ovary in the anatomic place of the left kidney. Dilatation and curettage was performed. Histology showed complex atypical endometrial hyperplasia. The patient underwent total abdominal hysterectomy with bilateral salpingo-oophorectomy, in an extremely interesting operation due to the multiple genitourinary anomalies. The uterus with a 6-centimeter uterine myoma and the adnexae were removed en block. Great effort was put into dissecting the left fallopian tube which arised from the cervix and via the rudimentary horn led to the left ectopic ovary that was located at the left kidneys' anatomic space. The patient recovered well and final histology was negative for malignancy. DISCUSSION: All necessary imaging examinations have to be scheduled prior to surgical intervention in order to give valuable anatomic information in cases of women diagnosed with Mullerian abnormalities.


Assuntos
Anormalidades Múltiplas/cirurgia , Coristoma/complicações , Endométrio/patologia , Histerectomia/métodos , Rim , Mioma/cirurgia , Ovário , Salpingo-Ooforectomia/métodos , Neoplasias Uterinas/cirurgia , Útero/anormalidades , Anormalidades Múltiplas/embriologia , Adulto , Colo do Útero/anormalidades , Tubas Uterinas/anormalidades , Feminino , Fertilização in vitro , Humanos , Hiperplasia , Infertilidade Feminina/etiologia , Infertilidade Feminina/terapia , Menorragia/etiologia , Metrorragia/etiologia , Ductos Paramesonéfricos/anormalidades , Ductos Paramesonéfricos/embriologia , Mioma/patologia , Pelve , Neoplasias Uterinas/patologia
9.
J Vet Med Sci ; 80(4): 557-567, 2018 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-29526868

RESUMO

Anti-Müllerian hormone (AMH) produced in the developing testis induces the regression of the Müllerian duct, which develops into the oviducts, uterus and upper vagina. In our true hermaphrodite mouse with an ovary on one side and a testis on the other (O/T), the oviduct and uterus are present only on the ovary side, and nothing derived from the Müllerian duct is present on the testis side. Here, we investigate the mechanism underlying the unilateral Müllerian duct regression and the mode of AMH signaling, by performing immunohistology, Western blotting, and organ culture analyses. The histological analysis revealed that during the start of the Müllerian duct regression, the duct in the O/T mice was clearly regressed on the AMH-positive testis side compared to the AMH-negative ovary side. The immunohistochemistry showed a diffuse immunoreaction of AMH in the interstitium surrounding the testis cord and boundary region between the testis and mesonephros, especially in the cranial portion. Western blotting revealed that the amount of AMH in the cranial half of the mesonephros was larger than that in the caudal half. AMH injected into the gonads in organ culture induced the regression of the Müllerian duct via the interstitium of the organ. These results suggest that AMH acts on the Müllerian duct in male mice by exuding into the interstitium surrounding the testis cord and infiltrating through the cranial region from the testis to the mesonephros.


Assuntos
Hormônio Antimülleriano/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ductos Paramesonéfricos/embriologia , Testículo/embriologia , Testículo/metabolismo , Animais , Hormônio Antimülleriano/genética , Feminino , Organismos Hermafroditas , Masculino , Camundongos , Receptores de Fatores de Crescimento Transformadores beta , Diferenciação Sexual
10.
J Pediatr Urol ; 14(2): 144-149, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29459133

RESUMO

BACKGROUND: Mullerian anomalies have a known association with renal agenesis yet, to date, there are no formal recommendations for screening women with certain renal anomalies for associated genital tract disorders. OBJECTIVE: The objective of this study is to review current data regarding the association between renal and Mullerian anomalies, and propose screening recommendations. STUDY DESIGN: A comprehensive review of the literature was performed to identify relevant articles using the keywords "unilateral renal agenesis," "renal anomalies," and "Mullerian anomalies." RESULTS: Over 30% of patients with unilateral renal agenesis have an associated Mullerian anomaly. However, diagnosis is frequently delayed in this population until after menarche when complications of retrograde menstruation with obstructive anomalies lead to significant problems including endometriosis, pelvic inflammatory disease, and infertility. No clear guidelines exist for communication among the antenatal sonographer, the obstetrician, the parents, and the child's pediatrician, which creates a barrier to effective screening and follow-up. Further, no current guidelines exist for screening women with certain renal anomalies for Mullerian anomalies. DISCUSSION: The complications of Mullerian anomalies are easily preventable if identified early. We propose new guidelines for education and screening for Mullerian anomalies in patients with unilateral renal agenesis (URA) and multicystic dysplastic kidney (MCDK) to guide providers, patients, and parents on proper identification and management (Table). CONCLUSIONS: Screening young women with URA and MCDK for Mullerian anomalies has the potential to prevent long-term complications from untreated obstructive malformations. Identification of unilateral renal agenesis on antenatal ultrasound must be clearly articulated with parents and the child's pediatrician so that proper screening can be performed before menarche. Pelvic sonography is a low-cost, high-yield screening tool to identify these anomalies.


Assuntos
Anormalidades Congênitas/diagnóstico , Nefropatias/congênito , Nefropatias/prevenção & controle , Rim/anormalidades , Ductos Paramesonéfricos/anormalidades , Triagem Neonatal/métodos , Guias de Prática Clínica como Assunto , Rim Único/diagnóstico , Anormalidades Congênitas/epidemiologia , Diagnóstico Precoce , Feminino , Humanos , Recém-Nascido , Nefropatias/diagnóstico , Nefropatias/epidemiologia , Masculino , Ductos Paramesonéfricos/embriologia , Medição de Risco , Rim Único/epidemiologia
11.
Wiley Interdiscip Rev Dev Biol ; 7(3): e310, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29350886

RESUMO

The Müllerian ducts are part of the embryonic urogenital system. They give rise to mature structures that serve a critical function in the transport and development of the oocyte and/or embryo. In most vertebrates, both sexes initially develop Müllerian ducts during embryogenesis, but they regress in males under the influence of testis-derived Anti-Müllerian Hormone (AMH). A number of regulatory factors have been shown to be essential for proper duct development, including Bmp and Wnt signaling molecules, together with homeodomain transcription factors such as PAX2 and LIM1. Later in development, the fate of the ducts diverges between males and females and is regulated by AMH and Wnt signaling molecules (duct regression in males) and Hox genes (duct patterning in females). Most of the genes and molecular pathways known to be involved in Müllerian duct development have been elucidated through animal models, namely, the mouse and chicken. In addition, genetic analysis of humans with reproductive tract disorders has further defined molecular mechanisms of duct formation and differentiation. However, despite our current understanding of Müllerian duct development, some questions remain to be answered at the molecular genetic level. This article is categorized under: Early Embryonic Development > Development to the Basic Body Plan.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Ductos Paramesonéfricos/embriologia , Diferenciação Sexual , Animais , Linhagem da Célula , Feminino , Humanos , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Ductos Paramesonéfricos/citologia , Ductos Paramesonéfricos/metabolismo , Via de Sinalização Wnt
12.
Eur J Obstet Gynecol Reprod Biol ; 212: 20-24, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28327376

RESUMO

OBJECTIVE: To evaluate whether offspring of women with müllerian anomalies are at an increased risk for long-term pediatric morbidity. STUDY DESIGN: A population-based cohort study compared the incidence of long-term (up to the age of 18 years for offspring) hospitalizations due to cardiovascular, endocrine, neurological, hematological, respiratory and urinary morbidity of offspring to mothers diagnosed with uterine anomalies. Deliveries occurred between the years 1991 and 2013 in a tertiary medical center. Multiple pregnancies, unknown gestational age, gestational age of less than 24 weeks, perinatal mortality, and fetuses with congenital malformations were excluded. Kaplan-Meier survival curves were used to compare cumulative morbidity incidence. Survival analysis for clustered data was performed for each major-system pediatric hospitalization. RESULTS: During the study period 253,808 deliveries met the inclusion criteria; 0.48% (n=1230) of which occurred in women diagnosed with müllerian anomalies. In the Kaplan-Meier survival curve, children born to mothers with müllerian anomalies did not have a significantly different cumulative incidence of any of the long-term pediatric morbidities, compared with the comparison group. In the multivariable survival analysis for clustered data, after adjustment of maternal clusters, maternal age, gestational age and birthweight, müllerian anomalies did not exhibit an independent association with long-term morbidities of the offspring. CONCLUSION: Müllerian anomalies do not appear to have an independent impact on long-term morbidity of the offspring.


Assuntos
Ductos Paramesonéfricos/anormalidades , Resultado da Gravidez , Útero/anormalidades , Adulto , Feminino , Idade Gestacional , Humanos , Recém-Nascido de Baixo Peso , Recém-Nascido Prematuro , Estimativa de Kaplan-Meier , Ductos Paramesonéfricos/embriologia , Gravidez , Estudos Retrospectivos , Estatísticas não Paramétricas , Vagina/anormalidades
13.
Proc Natl Acad Sci U S A ; 113(50): 14354-14359, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911779

RESUMO

The Müllerian duct develops into the oviduct, uterus, and vagina, all of which are quite distinct in their morphology and function. The epithelial fate of these female reproductive organs in developing mice is determined by factors secreted from the stroma; however, how stromal differentiation occurs in the female reproductive organs derived from the Müllerian duct is still unclear. In the present study, roles of retinoic acid (RA) signaling in developing female reproductive tracts were investigated. Retinol dehydrogenase 10 (RDH10) and aldehyde dehydrogenase family 1 subfamily A2 (ALDH1A2) mRNAs and proteins and transactivation activity of endogenous RA were found in the stroma of proximal Müllerian ducts and gradually decreased from the proximal to caudal regions in fetal mice. In organ-cultured Müllerian ducts, retinaldehyde or RA treatment induced uterine epithelial differentiation, defined as a layer of columnar epithelial cells negative for oviductal and vaginal epithelial markers. In contrast, inhibition of RA receptor (RAR) signaling induced vaginal epithelial differentiation, characterized as vaginal epithelial marker genes-positive stratified epithelium. Grafting experiments of the organ-cultured Müllerian duct revealed irreversible epithelial fate determination. Although RAR did not directly bind to the homeobox A10 (Hoxa10) promoter region, RA-RAR signaling stimulated Hoxa10 expression. Thus, RA-RAR signaling in the Müllerian duct determines the fate of stroma to form the future uterus and vagina.


Assuntos
Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Tretinoína/metabolismo , Útero/embriologia , Útero/metabolismo , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ductos Paramesonéfricos/citologia , Técnicas de Cultura de Órgãos , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Ácido Retinoico/antagonistas & inibidores , Retinal Desidrogenase , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional , Útero/citologia , Vagina/citologia , Vagina/embriologia , Vagina/metabolismo
14.
Stem Cells Dev ; 25(22): 1733-1741, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27595922

RESUMO

A fundamental understanding of prostate development and tissue homeostasis has the high potential to reveal mechanisms for prostate disease initiation and identify novel therapeutic approaches for disease prevention and treatment. Our current understanding of prostate lineage specification stems from the use of developmental model systems that rely upon the embryonic preprostatic urogenital sinus mesenchyme to induce the formation of mature prostate epithelial cells. It is unclear, however, how the urogenital sinus epithelium can derive both adult urethral glands and prostate epithelia. Furthermore, the vast disparity in disease initiation between these two glands highlights key developmental factors that predispose prostate epithelia to hyperplasia and cancer. In this study we demonstrate that the caudal Müllerian duct mesenchyme (CMDM) drives prostate epithelial differentiation and is a key determinant in cell lineage specification between urethral glands and prostate epithelia. Utilizing both human embryonic stem cells and mouse embryonic tissues, we document that the CMDM is capable of inducing the specification of androgen receptor, prostate-specific antigen, NKX3.1, and Hoxb13-positive prostate epithelial cells. These results help to explain key developmental differences between prostate and urethral gland differentiation, and implicate factors secreted by the caudal Müllerian duct as novel targets for prostate disease prevention and treatment.


Assuntos
Mesoderma/embriologia , Ductos Paramesonéfricos/embriologia , Organogênese , Próstata/embriologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Epitélio , Células-Tronco Embrionárias Humanas/citologia , Humanos , Masculino , Mesoderma/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ductos Paramesonéfricos/citologia , Próstata/citologia , Fatores de Transcrição/metabolismo , Uretra/citologia
15.
J Appl Toxicol ; 36(12): 1639-1650, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27143402

RESUMO

The Larval Amphibian Growth and Development Assay (LAGDA) is a globally harmonized test guideline developed by the U.S. Environmental Protection Agency in collaboration with Japan's Ministry of the Environment. The LAGDA was designed to evaluate apical effects of chronic chemical exposure on growth, thyroid-mediated amphibian metamorphosis and reproductive development. During the validation phase, two well-characterized endocrine-disrupting chemicals were tested to evaluate the performance of the initial assay design: xenoestrogen 4-tert-octylphenol (tOP) and xenoandrogen 17ß-trenbolone (TB). Xenopus laevis embryos were exposed, in flow-through conditions, to tOP (nominal concentrations: 0.0, 6.25, 12.5, 25 and 50 µg l-1 ) or TB (nominal concentrations: 0.0, 12.5, 25, 50 and 100 ng l-1 ) until 8 weeks post-metamorphosis, at which time growth measurements were taken, and histopathology assessments were made of the gonads, reproductive ducts, liver and kidneys. There were no effects on growth in either study and no signs of overt toxicity, sex reversal or gonad dysgenesis. Exposure to tOP caused a treatment-related decrease in circulating thyroxine and an increase in thyroid follicular cell hypertrophy and hyperplasia (25 and 50 µg l-1 ) during metamorphosis. Müllerian duct development was affected after exposure to both chemicals; tOP exposure caused dose-dependent maturation of oviducts in both male and female frogs, whereas TB exposure caused accelerated Müllerian duct regression in males and complete regression in >50% of the females in the 100 ng l-1 treatment. Based on these results, the LAGDA performed adequately to evaluate apical effects of chronic exposure to two endocrine-active compounds and is the first standardized amphibian multiple life stage toxicity test to date. Published 2016. This article is a U.S. Government work and is in the public domain in the USA.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Monitoramento Ambiental/métodos , Metamorfose Biológica/efeitos dos fármacos , Fenóis/toxicidade , Acetato de Trembolona/toxicidade , Animais , Bioensaio , Relação Dose-Resposta a Droga , Feminino , Larva , Masculino , Ductos Paramesonéfricos/efeitos dos fármacos , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/crescimento & desenvolvimento , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/embriologia , Glândula Tireoide/crescimento & desenvolvimento , Xenopus laevis
17.
Gen Comp Endocrinol ; 238: 96-104, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-26994582

RESUMO

The molecular signaling processes involved the differentiation of the Müllerian duct (MD) into the female reproductive tract, or oviduct, in non-mammalian vertebrates are not well understood. Studies in mammals and birds indicate that steroid hormones play a role in this process, as the embryonic MD has been shown to be vulnerable to exogenous estrogens and progestins and environmental endocrine disrupting contaminants. In a previous study, developmental treatment with an estrogen receptor α (ERα) agonist, 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT), induced significant enlargement of the MD in alligator embryos incubated at a male-producing temperature, which was not observed in embryos treated with an estrogen receptor ß (ERß) agonist, 7-bromo-2-(4-hydroxyphenyl)-1,3-benzoxazol-5-ol (WAY 200070), or with 17ß-estradiol (E2). In order to understand the role of estrogen signaling in female alligator oviduct development, we incubated eggs at a female-producing temperature and treated them with E2 and these ER selective agonists, PPT and WAY 200070, just prior to the thermosensitive window of sex determination. At stage 27, one stage prior to hatching, PPT induced significant enlargement of the MD with precocious development of secretory glands and connective tissue differentiation similar to characteristics of mature adult oviduct. PPT treatment in ovo increased mRNA expression of ERß, progesterone receptor, androgen receptor and insulin-like growth factor 1 in MD at stage 27, while expression of ERα was decreased. Neither WAY 200070 nor E2 treatment induced these effects seen in PPT-treated MD. The results of this study provide insight into the critical factors for healthy reproductive system formation in this sentinel species, although further investigation is needed to determine whether the observed phenomena are directly due to selective stimulation of ERα or related to some other aspect of PPT treatment.


Assuntos
Jacarés e Crocodilos/embriologia , Jacarés e Crocodilos/metabolismo , Receptor alfa de Estrogênio/agonistas , Genitália Feminina/embriologia , Animais , Embrião não Mamífero/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Feminino , Genitália Feminina/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Ductos Paramesonéfricos/efeitos dos fármacos , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Oxazóis/farmacologia , Fenóis/farmacologia , Pirazóis/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estados Unidos
18.
Gen Comp Endocrinol ; 229: 132-44, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26987287

RESUMO

Amphibian gonadal differentiation involves the action of sex steroids. Recent research indicates that the anti-Müllerian hormone (AMH) is involved in testicular development in some lower vertebrate species. For amphibians there is a lack of data on ontogenetic expression of the AMH receptor AMHR2/amhr2 and of progesterone receptors (PGRS/pgrs). Here we expand the knowledge on amphibian sex differentiation by characterizing ontogenetic mRNA levels of amh, amhr2, intracellular and membrane pgrs (ipgr and mpgr beta) and cytochrome P450 19a1 (cyp19a1) (ovarian marker) in the urogenital complex of the model species Xenopus (Silurana) tropicalis. Furthermore, we characterized the ontogenetic development of the Müllerian ducts (precursors of the female reproductive tract) histologically. The developmental period investigated spanned from beginning of gonadal differentiation, Nieuwkoop and Faber (NF) stage 51, to 4weeks post-metamorphosis. The Müllerian ducts were first observed at NF 64 in both sexes. Male-enhanced amh mRNA levels from NF 53/54 to 6days post-metamorphosis and female-enhanced cyp19a1 levels from NF 53 to 4weeks post-metamorphosis were noted. The sexually dimorphic mRNA level profile was more distinct for amh than for cyp19a1. The pgrs mRNA levels increased over the studied period and showed no sex differences. At later developmental stages, the amhr2 mRNA level was increased in putative females compared with males. Our findings suggest that AMH has a role in gonadal differentiation in X. tropicalis. We propose relative gonadal amh mRNA level as a testicular marker during early gonadal development in amphibians.


Assuntos
Hormônio Antimülleriano/metabolismo , Ductos Paramesonéfricos/embriologia , Diferenciação Sexual/genética , Xenopus/metabolismo , Animais , Diferenciação Celular , Feminino , Masculino , Receptores de Peptídeos , Receptores de Fatores de Crescimento Transformadores beta
19.
Reprod Biomed Online ; 32(4): 420-6, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26856455

RESUMO

Dapper antagonist of catenin-1 (DACT1) plays an important role in embryogenesis and organogenesis of the female reproductive tract in mouse models. The aim of this study was to investigate the association between DACT1 mutations and human Müllerian duct anomalies (MDA). One hundred clinically well-defined Chinese Han patients with MDA and 200 healthy controls were recruited in this study. All four exons coding for DACT1 were amplified and sequenced. A missense mutation (c.G1084A, p.V362M) was identified in a patient who had a didelphic uterus and was absent from the control group. This variant changed the hydrophilicity of the amino acid residue and was predicted to be deleterious to the structure and function of DACT1 protein. The data indicate that the p.V362M mutation of DACT1 may be an underlying cause of MDA.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Ductos Paramesonéfricos/anormalidades , Proteínas Nucleares/genética , Adulto , China , Análise Mutacional de DNA , Feminino , Humanos , Ductos Paramesonéfricos/embriologia , Mutação de Sentido Incorreto , Via de Sinalização Wnt
20.
Reprod Biomed Online ; 32(4): 388-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26896429

RESUMO

Homeobox A10 (HOXA10) and empty spiracles homeobox 2 (EMX2) are two transcription factors necessary for female Müllerian duct differentiation and development. They are thought to play important roles in embryo implantation in mice and humans. The EMX2 gene is a known direct target of HOXA10 in the reproductive tract. Human TENM1 is directly regulated by EMX2 and is expressed during embryonic pattern formation and morphogenesis. This study aimed to investigate expression patterns of HOXA10, EMX2 and TENM1 in the mid-secretory endometrium of infertile patients with a Müllerian duct anomaly causing a partially septate uterus. Thirteen mid-secretory endometrial tissue samples were collected from women with partially septate uteri and 12 from women with normal uteri as controls. Expression levels of HOXA10, EMX2 and TENM1 mRNA and protein in the mid-secretory endometrium of infertile patients and controls were measured by quantitative reverse transcription polymerase chain reaction and western blotting. Compared with controls, mRNA and protein expression levels of HOXA10 decreased significantly (P < 0.01), whereas EMX2 and TENM1 increased dramatically in patients with Müllerian duct anomaly (P < 0.001). Changes in HOXA10, EMX2, and TENM1 expression levels might act in infertile women with Müllerian duct anomaly to cause a partially septate uterus.


Assuntos
Proteínas de Homeodomínio/metabolismo , Infertilidade/metabolismo , Fase Luteal , Ductos Paramesonéfricos/anormalidades , Proteínas do Tecido Nervoso/metabolismo , Tenascina/metabolismo , Fatores de Transcrição/metabolismo , Western Blotting , Feminino , Proteínas Homeobox A10 , Proteínas de Homeodomínio/genética , Humanos , Infertilidade/genética , Ductos Paramesonéfricos/embriologia , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tenascina/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA