Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530552

RESUMEN

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.


Asunto(s)
Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Proteínas Hedgehog/metabolismo , Organogénesis , Transducción de Señal , Útero/embriología , Útero/metabolismo , Animales , Biomarcadores , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Modelos Biológicos , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Organogénesis/genética
2.
BMC Genomics ; 21(1): 688, 2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33008304

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Conductos Paramesonéfricos/metabolismo , Transcriptoma , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Embrión de Pollo , Femenino , Conductos Paramesonéfricos/embriología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
J Toxicol Environ Health A ; 83(2): 66-81, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-32077375

RESUMEN

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.


Asunto(s)
Compuestos de Bencidrilo/metabolismo , Compuestos de Bencidrilo/toxicidad , Fenoles/metabolismo , Fenoles/toxicidad , Ácidos Ftálicos/química , Procesos de Determinación del Sexo/efectos de los fármacos , Animales , Compuestos de Bencidrilo/química , Embrión de Pollo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Conductos Paramesonéfricos/efectos de los fármacos , Conductos Paramesonéfricos/embriología , Ovario/efectos de los fármacos , Ovario/embriología , Fenoles/química , Ácidos Ftálicos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Testículo/efectos de los fármacos , Testículo/embriología
4.
Proc Natl Acad Sci U S A ; 113(50): 14354-14359, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911779

RESUMEN

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.


Asunto(s)
Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Tretinoina/metabolismo , Útero/embriología , Útero/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Conductos Paramesonéfricos/citología , Técnicas de Cultivo de Órganos , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Retinal-Deshidrogenasa , Transducción de Señal/efectos de los fármacos , Activación Transcripcional , Útero/citología , Vagina/citología , Vagina/embriología , Vagina/metabolismo
5.
G Chir ; 39(4): 245-247, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30039793

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/cirugía , Coristoma/complicaciones , Endometrio/patología , Histerectomía/métodos , Riñón , Mioma/cirugía , Ovario , Salpingooforectomía/métodos , Neoplasias Uterinas/cirugía , Útero/anomalías , Anomalías Múltiples/embriología , Adulto , Cuello del Útero/anomalías , Trompas Uterinas/anomalías , Femenino , Fertilización In Vitro , Humanos , Hiperplasia , Infertilidad Femenina/etiología , Infertilidad Femenina/terapia , Menorragia/etiología , Metrorragia/etiología , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/embriología , Mioma/patología , Pelvis , Neoplasias Uterinas/patología
6.
Dev Biol ; 400(2): 224-36, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25684667

RESUMEN

DMRT1 is a conserved transcription factor with a central role in gonadal sex differentiation. In all vertebrates studied, DMRT1 plays an essential function in testis development and/or maintenance. No studies have reported a role for DMRT1 outside the gonads. Here, we show that DMRT1 is expressed in the paired Müllerian ducts in the chicken embryo, where it is required for duct formation. DMRT1 mRNA and protein are expressed in the early forming Müllerian ridge, and in cells undergoing an epithelial to mesenchyme transition during duct morphogenesis. RNAi-mediated knockdown of DMRT1 in ovo causes a greatly reduced mesenchymal layer, which blocks caudal extension of the duct luminal epithelium. Critical markers of Müllerian duct formation in mammals, Pax2 in the duct epithelium and Wnt4 in the mesenchyme, are conserved in chicken and their expression disrupted in DMRT1 knockdown ducts. We conclude that DMRT1 is required for the early steps of Müllerian duct development. DMRT1 regulates Müllerian ridge and mesenchyme formation and its loss blocks caudal extension of the duct. While DMRT1 plays an important role during testis development and maintenance in many vertebrate species, this is the first report showing a requirement for DMRT1 in Müllerian duct development.


Asunto(s)
Proteínas Aviares/metabolismo , Conductos Paramesonéfricos/embriología , Factores de Transcripción/metabolismo , Animales , Embrión de Pollo , Electroporación , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Mesodermo/metabolismo , Conductos Paramesonéfricos/metabolismo , Oviductos/embriología , Organismos Libres de Patógenos Específicos , Testículo/embriología , Factores de Transcripción/genética , Vertebrados/embriología , Vertebrados/metabolismo
7.
Reprod Biomed Online ; 32(4): 420-6, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26856455

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Conductos Paramesonéfricos/anomalías , Proteínas Nucleares/genética , Adulto , China , Análisis Mutacional de ADN , Femenino , Humanos , Conductos Paramesonéfricos/embriología , Mutación Missense , Vía de Señalización Wnt
8.
Reprod Biomed Online ; 32(4): 388-93, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26896429

RESUMEN

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.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Infertilidad/metabolismo , Fase Luteínica , Conductos Paramesonéfricos/anomalías , Proteínas del Tejido Nervioso/metabolismo , Tenascina/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Femenino , Proteínas Homeobox A10 , Proteínas de Homeodominio/genética , Humanos , Infertilidad/genética , Conductos Paramesonéfricos/embriología , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tenascina/genética , Factores de Transcripción/genética
9.
Gen Comp Endocrinol ; 229: 132-44, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26987287

RESUMEN

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.


Asunto(s)
Hormona Antimülleriana/metabolismo , Conductos Paramesonéfricos/embriología , Diferenciación Sexual/genética , Xenopus/metabolismo , Animales , Diferenciación Celular , Femenino , Masculino , Receptores de Péptidos , Receptores de Factores de Crecimiento Transformadores beta
10.
Gen Comp Endocrinol ; 238: 96-104, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-26994582

RESUMEN

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.


Asunto(s)
Caimanes y Cocodrilos/embriología , Caimanes y Cocodrilos/metabolismo , Receptor alfa de Estrógeno/agonistas , Genitales Femeninos/embriología , Animales , Embrión no Mamífero/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Genitales Femeninos/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Conductos Paramesonéfricos/efectos de los fármacos , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Oxazoles/farmacología , Fenoles/farmacología , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estados Unidos
11.
J Appl Toxicol ; 36(12): 1639-1650, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27143402

RESUMEN

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.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Monitoreo del Ambiente/métodos , Metamorfosis Biológica/efectos de los fármacos , Fenoles/toxicidad , Acetato de Trembolona/toxicidad , Animales , Bioensayo , Relación Dosis-Respuesta a Droga , Femenino , Larva , Masculino , Conductos Paramesonéfricos/efectos de los fármacos , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/crecimiento & desarrollo , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/embriología , Glándula Tiroides/crecimiento & desarrollo , Xenopus laevis
12.
Dev Biol ; 386(1): 227-36, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24362065

RESUMEN

A key event during mammalian sexual development is regression of the Müllerian ducts (MDs) in the bipotential urogenital ridges (UGRs) of fetal males, which is caused by the expression of Müllerian inhibiting substance (MIS) in the Sertoli cells of the differentiating testes. The paracrine signaling mechanisms involved in MD regression are not completely understood, particularly since the receptor for MIS, MISR2, is expressed in the mesenchyme surrounding the MD, but regression occurs in both the epithelium and mesenchyme. Microarray analysis comparing MIS signaling competent and Misr2 knockout embryonic UGRs was performed to identify secreted factors that might be important for MIS-mediated regression of the MD. A seven-fold increase in the expression of Wif1, an inhibitor of WNT/ß-catenin signaling, was observed in the Misr2-expressing UGRs. Whole mount in situ hybridization of Wif1 revealed a spatial and temporal pattern of expression consistent with Misr2 during the window of MD regression in the mesenchyme surrounding the MD epithelium that was absent in both female UGRs and UGRs knocked out for Misr2. Knockdown of Wif1 expression in male UGRs by Wif1-specific siRNAs beginning on embryonic day 13.5 resulted in MD retention in an organ culture assay, and exposure of female UGRs to added recombinant human MIS induced Wif1 expression in the MD mesenchyme. Knockdown of Wif1 led to increased expression of ß-catenin and its downstream targets TCF1/LEF1 in the MD mesenchyme and to decreased apoptosis, resulting in partial to complete retention of the MD. These results strongly suggest that WIF1 secretion by the MD mesenchyme plays a role in MD regression in fetal males.


Asunto(s)
Hormona Antimülleriana/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Conductos Paramesonéfricos/embriología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Femenino , Perfilación de la Expresión Génica , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Conductos Paramesonéfricos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/metabolismo , Células de Sertoli/metabolismo , Transducción de Señal , Factores de Tiempo
13.
Development ; 138(10): 1967-75, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490063

RESUMEN

During male sexual differentiation, the transforming growth factor-ß (TGF-ß) signaling molecule anti-Müllerian hormone (AMH; also known as Müllerian inhibiting substance, MIS) is secreted by the fetal testes and induces regression of the Müllerian ducts, the primordia of the female reproductive tract organs. Currently, the molecular identity of downstream events regulated by the AMH signaling pathway remains unclear. We found that male-specific Wnt4 expression in mouse Müllerian duct mesenchyme depends upon AMH signaling, implicating the WNT pathway as a downstream mediator of Müllerian duct regression. Inactivation of ß-catenin, a mediator of the canonical WNT pathway, did not affect AMH signaling activation in the Müllerian duct mesenchyme, but did block Müllerian duct regression. These data suggest that ß-catenin mediates AMH signaling for Müllerian duct regression during male sexual differentiation.


Asunto(s)
Conductos Paramesonéfricos/embriología , Diferenciación Sexual/fisiología , Testículo/embriología , beta Catenina/fisiología , Animales , Hormona Antimülleriana/fisiología , Secuencia de Bases , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Conductos Paramesonéfricos/metabolismo , Embarazo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Diferenciación Sexual/genética , Transducción de Señal , Testículo/metabolismo , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/fisiología , Proteína Wnt4 , beta Catenina/deficiencia , beta Catenina/genética
14.
Poult Sci ; 93(4): 953-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24706973

RESUMEN

Sex of birds is genetically determined by the inheritance of sex chromosomes (ZZ for male and ZW for female), and the Z-linked gene named doublesex and mab-3 related transcription factor 1 (DMRT1) is a candidate sex-determining gene in avian species. However, the mechanisms underlying sex determination in birds are not yet understood, and the expression patterns of the DMRT1 protein in urogenital tissues have not been identified. In the current study, we used immunohistochemistry to investigate the detailed expression patterns of the DMRT1 protein in the urogenital systems (including Müllerian ducts) in male and female chicken embryos throughout embryonic development. Gonadal somatic cells in the male indifferent gonads showed stronger expressions of DMRT1 compared with those in the female indifferent gonads well before the presumptive period of the sex determination, and Sertoli cells forming testicular cords expressed DMRT1 in the testes after sex determination. Germ cells expressed DMRT1 equally in males and females after sex determination. The expression was continuous in males, but in females it gradually disappeared from the germ cells in the central part of the cortex of the left ovary toward both edges. The DMRT1 was also detected in the tubal ridge, which is a precursor of the Müllerian duct, and at the mesenchyme and outermost coelomic epithelium of the Müllerian duct in both sexes. Strong expression was observed in the males, but it was restricted to coelomic epithelium after the regression of the duct started. Thus, we observed the detailed spatiotemporal expression patterns of DMRT1 in the developing chicken urogenital systems throughout embryonic development, suggesting its various roles in the development of urogenital tissues in the chicken embryo.


Asunto(s)
Embrión de Pollo/embriología , Pollos/genética , Conductos Paramesonéfricos/embriología , Ovario/embriología , Testículo/embriología , Factores de Transcripción/genética , Animales , Femenino , Inmunohistoquímica/veterinaria , Masculino , Conductos Paramesonéfricos/citología , Conductos Paramesonéfricos/metabolismo , Ovario/citología , Ovario/metabolismo , Procesos de Determinación del Sexo , Testículo/citología , Testículo/metabolismo , Factores de Transcripción/metabolismo
15.
Toxicol Sci ; 199(2): 210-226, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38526210

RESUMEN

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.


Asunto(s)
Coturnix , Etinilestradiol , Regulación del Desarrollo de la Expresión Génica , Conductos Paramesonéfricos , Testículo , Animales , Masculino , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/embriología , Testículo/patología , Coturnix/embriología , Coturnix/genética , Etinilestradiol/toxicidad , Conductos Paramesonéfricos/efectos de los fármacos , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/anomalías , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Feminización/inducido químicamente , Feminización/genética
16.
J Anat ; 220(4): 363-71, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22256858

RESUMEN

Unresolved questions remain concerning the derivation of the vagina with respect to the relative contributions from the Müllerian ducts, the urogenital sinus, and the Wolffian ducts. Recent molecular and cellular studies in rodents have opened up a large gap between the level of understanding of vaginal development in mice and understanding of human vaginal development, which is based on histology. To compare the findings in mice with human vaginal development and to address this gap, we analysed molecular characteristics of the urogenital sinus, Wolffian ducts, and Müllerian ducts in 8-14-week-old human specimens using immunohistochemical methods. The monoclonal antibodies used were directed against cytokeratin (CK) 14, CK19, vimentin, laminin, p63, E-cadherin, caspase-3, Ki67, HOX A13, and BMP-4. The immunohistochemical analysis revealed that, during weeks 8-9, the epithelium of the Müllerian ducts became positive for p63 as p63-positive cells that originated from the sinus epithelium reached the caudal tip of the fused Müllerian ducts via the Wolffian ducts. The lumen of the fused Müllerian ducts was closed by an epithelial plug that contained both vimentin-positive and vimentin-negative cells. Subsequently, the resulting epithelial tube enlarged by proliferation of basal p63-positive cells. The first signs of squamous differentiation were detected during week 14, with the appearance of CK14-positive cells. According to our results, all three components, namely, the urogenital sinus, Wolffian ducts, and Müllerian ducts, interacted during the formation of the human vagina. The sinus epithelium provided p63-positive cells, the Wollfian ducts acted as a 'transporter', and the Müllerian ducts contributed the guiding structure for the vaginal anlagen. Epithelial differentiation began at the end of the period studied and extended in a caudo-cranial direction. The present study is one of the first to provide up-to-date molecular correlates for human vaginal development that can be compared with the results of cell biological studies in rodents.


Asunto(s)
Vagina/embriología , Epitelio/metabolismo , Femenino , Humanos , Inmunohistoquímica , Conductos Paramesonéfricos/anatomía & histología , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/fisiología , Sistema Urogenital/anatomía & histología , Sistema Urogenital/embriología , Sistema Urogenital/fisiología , Conductos Mesonéfricos/anatomía & histología , Conductos Mesonéfricos/embriología , Conductos Mesonéfricos/fisiología
17.
Differentiation ; 82(3): 117-26, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21612855

RESUMEN

In mammals, the female reproductive tract (FRT) develops from a pair of paramesonephric or Müllerian ducts (MDs), which arise from coelomic epithelial cells of mesodermal origin. During development, the MDs undergo a dynamic morphogenetic transformation from simple tubes consisting of homogeneous epithelium and surrounding mesenchyme into several distinct organs namely the oviduct, uterus, cervix and vagina. Following the formation of anatomically distinctive organs, the uniform MD epithelium (MDE) differentiates into diverse epithelial cell types with unique morphology and functions in each organ. Classic tissue recombination studies, in which the epithelium and mesenchyme isolated from the newborn mouse FRT were recombined, have established that the organ specific epithelial cell fate of MDE is dictated by the underlying mesenchyme. The tissue recombination studies have also demonstrated that there is a narrow developmental window for the epithelial cell fate determination in MD-derived organs. Accordingly, the developmental plasticity of epithelial cells is mostly lost in mature FRT. If the signaling that controls epithelial differentiation is disrupted at the critical developmental stage, the cell fate of MD-derived epithelial tissues will be permanently altered and can result in epithelial lesions in adult life. A disruption of signaling that maintains epithelial cell fate can also cause epithelial lesions in the FRT. In this review, the pathogenesis of cervical/vaginal adenoses and uterine squamous metaplasia is discussed as examples of such incidences.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Conductos Paramesonéfricos/citología , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Animales , Cuello del Útero/anomalías , Cuello del Útero/citología , Cuello del Útero/embriología , Dietilestilbestrol/efectos adversos , Células Epiteliales/metabolismo , Estrógenos no Esteroides/efectos adversos , Femenino , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Metaplasia , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Organogénesis , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Útero/citología , Útero/embriología , Vagina/anomalías , Vagina/citología , Vagina/embriología
18.
Ginecol Obstet Mex ; 80(10): 663-7, 2012 Oct.
Artículo en Español | MEDLINE | ID: mdl-23240231

RESUMEN

The Mayer-Rokitansky-Kuster-Hauser is a rare congenital anomaly characterized by lack of vaginal and uterine development variable and normal ovaries. It results from agenesis or hypoplasia Müller duct system. Cervicovaginal agenesis as part of the complex syndrome, is even rarer. We report two cases: adolescent patient with primary amenorrhea, cervicovaginal agenesis and chronic pelvic pain, and a 28-year-old patient with primary amenorrhea, congenital absence of uterus and vagina.


Asunto(s)
Anomalías Múltiples/patología , Amenorrea/etiología , Trastornos del Desarrollo Sexual 46, XX , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/embriología , Anomalías Múltiples/epidemiología , Anomalías Múltiples/cirugía , Adolescente , Adulto , Anomalías Congénitas , Endometriosis/etiología , Femenino , Humanos , Histerectomía , Incidencia , Riñón/anomalías , Riñón/diagnóstico por imagen , Riñón/embriología , Riñón/patología , Riñón/cirugía , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/diagnóstico por imagen , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/patología , Conductos Paramesonéfricos/cirugía , Dolor Pélvico/etiología , Fenotipo , Somitos/anomalías , Somitos/diagnóstico por imagen , Somitos/embriología , Somitos/patología , Somitos/cirugía , Columna Vertebral/anomalías , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/embriología , Columna Vertebral/patología , Columna Vertebral/cirugía , Estructuras Creadas Quirúrgicamente , Ultrasonografía , Útero/anomalías , Útero/diagnóstico por imagen , Útero/embriología , Útero/patología , Útero/cirugía , Vagina/anomalías , Vagina/diagnóstico por imagen , Vagina/embriología , Vagina/patología , Vagina/cirugía
19.
Differentiation ; 80(2-3): 99-105, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20638775

RESUMEN

The developmental origin of vaginal epithelium has been controversial for nearly a century, with speculation that vaginal epithelium originates from the Müllerian duct, Wolffian duct, and/or urogenital sinus. None of these possibilities have been definitively proven or disproven by direct scientific data. To define precisely the origin of vaginal epithelium, epithelial cells of the Müllerian duct, Wolffian duct, or urogenital sinus were fluorescently labeled in mouse embryos by crossing tdTomato-EGFP dual-reporter transgenic mice with transgenic mouse lines that express Cre-recombinase in each type of epithelium. In embryos and newborn mice, the vagina consisted of fused Müllerian ducts plus the sinus vagina of urogenital sinus origin. However, the proportion of the sinus vagina was significantly reduced as the Müllerian vagina grew caudally. By postpartum day 7, the Müllerian vagina extended to the caudal end of the body, whereas the sinus vagina remained only at the junction between the vagina and perineal skin. As the vagina opened in puberty, urogenital sinus epithelium was detected only in the vulva, but not in the vagina. Additionally, from embryo to adult stages, residual Wolffian duct epithelium was present in the dorsolateral stromal wall of the vagina, but not within vaginal or vulvar epithelium. In conclusion, adult mouse vaginal epithelium is derived solely from Müllerian duct epithelium.


Asunto(s)
Vagina/embriología , Animales , Epitelio/embriología , Femenino , Técnica del Anticuerpo Fluorescente , Ratones , Microscopía Fluorescente , Conductos Paramesonéfricos/embriología
20.
Dev Biol ; 325(2): 351-62, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19013450

RESUMEN

In vertebrates, the Müllerian duct elongates along the Wolffian duct, a mesonephric structure that is required for Müllerian duct formation. Recently, several genes required for initial Müllerian duct formation have been identified. However, the precise mechanism of Müllerian duct elongation remains to be elucidated. In this study, we investigated dynamic morphological changes in the elongating Müllerian duct in rat urogenital ridges in organ culture manipulated by microincision and/or chemical inhibitors. Mechanical division of the developing Müllerian duct showed that epithelial cells of the Müllerian duct actively migrate along the anterior-posterior axis independent of the proliferative expansion of the anterior portion of the duct. We found that the PI3K/AKT signaling pathway is activated in the Müllerian duct epithelium and is required for elongation of the tip of the duct; however, migration of Müllerian duct epithelial cells proximal to the tip remains intact when PI3K/AKT is inactivated. Although much is known about the molecular and cellular mechanisms leading to Müllerian duct regression, the present findings provide a fuller understanding of the mechanisms contributing to Müllerian duct formation and to the general process of early tubulogenesis.


Asunto(s)
Movimiento Celular , Conductos Paramesonéfricos/embriología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proliferación Celular , Cromonas/farmacología , Activación Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Morfolinas/farmacología , Conductos Paramesonéfricos/citología , Conductos Paramesonéfricos/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Técnicas de Cultivo de Tejidos , Conductos Mesonéfricos/citología , Conductos Mesonéfricos/embriología , Conductos Mesonéfricos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA