Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Am J Hum Genet ; 108(2): 337-345, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33434492

RESUMEN

Mayer-Rokitansky-Küster-Hauser syndrome (MRKHS) is associated with congenital absence of the uterus, cervix, and the upper part of the vagina; it is a sex-limited trait. Disrupted development of the Müllerian ducts (MD)/Wölffian ducts (WD) through multifactorial mechanisms has been proposed to underlie MRKHS. In this study, exome sequencing (ES) was performed on a Chinese discovery cohort (442 affected subjects and 941 female control subjects) and a replication MRKHS cohort (150 affected subjects of mixed ethnicity from North America, South America, and Europe). Phenotypic follow-up of the female reproductive system was performed on an additional cohort of PAX8-associated congenital hypothyroidism (CH) (n = 5, Chinese). By analyzing 19 candidate genes essential for MD/WD development, we identified 12 likely gene-disrupting (LGD) variants in 7 genes: PAX8 (n = 4), BMP4 (n = 2), BMP7 (n = 2), TBX6 (n = 1), HOXA10 (n = 1), EMX2 (n = 1), and WNT9B (n = 1), while LGD variants in these genes were not detected in control samples (p = 1.27E-06). Interestingly, a sex-limited penetrance with paternal inheritance was observed in multiple families. One additional PAX8 LGD variant from the replication cohort and two missense variants from both cohorts were revealed to cause loss-of-function of the protein. From the PAX8-associated CH cohort, we identified one individual presenting a syndromic condition characterized by CH and MRKHS (CH-MRKHS). Our study demonstrates the comprehensive utilization of knowledge from developmental biology toward elucidating genetic perturbations, i.e., rare pathogenic alleles involving the same loci, contributing to human birth defects.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Anomalías Congénitas/genética , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/crecimiento & desarrollo , Mutación , Conductos Mesonéfricos/crecimiento & desarrollo , Adulto , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 7/genética , Codón sin Sentido , Femenino , Estudios de Asociación Genética , Pleiotropía Genética , Proteínas Homeobox A10/genética , Proteínas de Homeodominio/genética , Humanos , Factor de Transcripción PAX8/genética , Herencia Paterna , Penetrancia , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Proteínas Wnt/genética , Conductos Mesonéfricos/anomalías
2.
Biol Reprod ; 106(1): 9-23, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34494091

RESUMEN

The vertebrate female reproductive tract has undergone considerable diversification over evolution, having become physiologically adapted to different reproductive strategies. This review considers the female reproductive tract from the perspective of evolutionary developmental biology (evo-devo). Very little is known about how the evolution of this organ system has been driven at the molecular level. In most vertebrates, the female reproductive tract develops from paired embryonic tubes, the Müllerian ducts. We propose that formation of the Müllerian duct is a conserved process that has involved co-option of genes and molecular pathways involved in tubulogenesis in the adjacent mesonephric kidney and Wolffian duct. Downstream of this conservation, genetic regulatory divergence has occurred, generating diversity in duct structure. Plasticity of the Hox gene code and wnt signaling, in particular, may underlie morphological variation of the uterus in mammals, and evolution of the vagina. This developmental plasticity in Hox and Wnt activity may also apply to other vertebrates, generating the morphological diversity of female reproductive tracts evident today.


Asunto(s)
Evolución Biológica , Biología Evolutiva , Genitales Femeninos/crecimiento & desarrollo , Animales , Estrógenos , Trompas Uterinas/crecimiento & desarrollo , Femenino , Expresión Génica , Genes Homeobox , Genitales Femeninos/anatomía & histología , Humanos , Morfogénesis/genética , Morfogénesis/fisiología , Conductos Paramesonéfricos/crecimiento & desarrollo , Útero/crecimiento & desarrollo , Vertebrados , Vía de Señalización Wnt
3.
Differentiation ; 118: 34-40, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33707128

RESUMEN

The development of the female reproductive tract can be divided into three parts consisting of Müllerian duct organogenesis, pre-sexual maturation organ development, and post-sexual maturation hormonal regulation. In primates, Müllerian duct organogenesis proceeds in an estrogen independent fashion based on transcriptional pathways that are suppressed in males by the presence of AMH and SRY. However, clinical experience indicates that exposure to xenoestrogens such as diethylstilbestrol (DES) during critical periods including late organogenesis and pre-sexual maturational development can have substantial effects on uterine morphology, and confer increased risk of disease states later in life. Recent evidence has demonstrated that these effects are in part due to epigenetic regulation of gene expression, both in the form of aberrant CpG methylation, and accompanying histone modifications. While xenoestrogens and selective estrogen receptor modulators (SERMS) both can induce non-canonical binding confirmations in estrogen receptors, the primate specific fetal estrogens Estriol and Estetrol may act in a similar fashion to alter gene expression through tissue specific epigenetic modulation.


Asunto(s)
Metilación de ADN/genética , Estrógenos/genética , Genitales Femeninos/crecimiento & desarrollo , Organogénesis/genética , Animales , Metilación de ADN/efectos de los fármacos , Dietilestilbestrol/farmacología , Epigénesis Genética/genética , Estradiol/metabolismo , Estrógenos/metabolismo , Femenino , Genitales Femeninos/metabolismo , Humanos , Conductos Paramesonéfricos/efectos de los fármacos , Conductos Paramesonéfricos/crecimiento & desarrollo , Conductos Paramesonéfricos/metabolismo , Organogénesis/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Útero/efectos de los fármacos , Útero/crecimiento & desarrollo
4.
Differentiation ; 110: 49-63, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31622789

RESUMEN

The study of male and female reproductive tract development requires expertise in two separate disciplines, developmental biology and endocrinology. For ease of experimentation and economy, the mouse has been used extensively as a model for human development and pathogenesis, and for the most part similarities in developmental processes and hormone action provide ample justification for the relevance of mouse models for human reproductive tract development. Indeed, there are many examples describing the phenotype of human genetic disorders that have a reasonably comparable phenotype in mice, attesting to the congruence between mouse and human development. However, anatomic, developmental and endocrinologic differences exist between mice and humans that (1) must be appreciated and (2) considered with caution when extrapolating information between all animal models and humans. It is critical that the investigator be aware of both the similarities and differences in organogenesis and hormone action within male and female reproductive tracts so as to focus on those features of mouse models with clear relevance to human development/pathology. This review, written by a team with extensive expertise in the anatomy, developmental biology and endocrinology of both mouse and human urogenital tracts, focusses upon the significant human/mouse differences, and when appropriate voices a cautionary note regarding extrapolation of mouse models for understanding development of human male and female reproductive tracts.


Asunto(s)
Epitelio/crecimiento & desarrollo , Genitales Femeninos/crecimiento & desarrollo , Conductos Paramesonéfricos/crecimiento & desarrollo , Útero/crecimiento & desarrollo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Ratones , Organogénesis/fisiología
5.
Hum Reprod ; 34(6): 1117-1125, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31111890

RESUMEN

STUDY QUESTION: Is there an increased prevalence of male microchimerism in women with Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome, as evidence of fetal exposure to blood and anti-Müllerian hormone (AMH) from a (vanished) male co-twin resulting in regression of the Müllerian duct derivatives? SUMMARY ANSWER: Predominant absence of male microchimerism in adult women with MRKH syndrome does not support our hypothesis that intrauterine blood exchange with a (vanished) male co-twin is the pathophysiological mechanism. WHAT IS KNOWN ALREADY: The etiology of MRKH is unclear. Research on the phenotype analogous condition in cattle (freemartinism) has yielded the hypothesis that Müllerian duct development is inhibited by exposure to AMH in utero. In cattle, the male co-twin has been identified as the source for AMH, which is transferred via placental blood exchange. In human twins, a similar exchange of cellular material has been documented by detection of chimerism, but it is unknown whether this has clinical consequences. STUDY DESIGN, SIZE, DURATION: An observational case-control study was performed to compare the presence of male microchimerism in women with MRKH syndrome and control women. Through recruitment via the Dutch patients' association of women with MRKH (comprising 300 members who were informed by email or regular mail), we enrolled 96 patients between January 2017 and July 2017. The control group consisted of 100 women who reported never having been pregnant. PARTICIPANTS/MATERIALS, SETTING, METHODS: After written informed consent, peripheral blood samples were obtained by venipuncture, and genomic DNA was extracted. Male microchimerism was detected by Y-chromosome-specific real-time quantitative PCR, with use of DYS14 marker. Possible other sources for microchimerism, for example older brothers, were evaluated using questionnaire data. MAIN RESULTS AND THE ROLE OF CHANCE: The final analysis included 194 women: 95 women with MRKH syndrome with a mean age of 40.9 years and 99 control women with a mean age of 30.2 years. In total, 54 women (56.8%) were identified as having typical MRKH syndrome, and 41 women (43.2%) were identified as having atypical MRKH syndrome (when extra-genital malformations were present). The prevalence of male microchimerism was significantly higher in the control group than in the MRKH group (17.2% versus 5.3%, P = 0.009). After correcting for age, women in the control group were 5.8 times more likely to have male microchimerism (odds ratio 5.84 (CI 1.59-21.47), P = 0.008). The mean concentration of male microchimerism in the positive samples was 56.0 male genome equivalent per 1 000 000 cells. The prevalence of male microchimerism was similar in women with typical MRKH syndrome and atypical MRKH syndrome (5.6% versus 4.9%, P = 0.884). There were no differences between women with or without microchimerism in occurrence of alternative sources of XY cells, such as older brothers, previous blood transfusion, or history of sexual intercourse. LIMITATIONS, REASON FOR CAUTION: We are not able to draw definitive conclusions regarding the occurrence of AMH exchange during embryologic development in women with MRKH syndrome. Our subject population includes all adult women and therefore is reliant on long-term prevalence of microchimerism. Moreover, we have only tested blood, and, theoretically, the cells may have grafted anywhere in the body during development. It must also be considered that the exchange of AMH may occur without the transfusion of XY cells and therefore cannot be discovered by chimerism detection. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to test the theory that freemartinism causes the MRKH syndrome in humans. The study aimed to test the presence of male microchimerism in women with MRKH syndrome as a reflection of early fetal exposure to blood and AMH from a male (vanished) co-twin. We found that male microchimerism was only present in 5.3% of the women with MRKH syndrome, a significantly lower percentage than in the control group (17.2%). Our results do not provide evidence for an increased male microchimerism in adult women with MRKH as a product of intrauterine blood exchange. However, the significant difference in favor of the control group is of interest to the ongoing discussion on microchimeric cell transfer and the possible sources of XY cells. STUDY FUNDING/COMPETING INTEREST(S): None. TRIAL REGISTRATION NUMBER: Dutch trial register, NTR5961.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Quimerismo , Anomalías Congénitas/genética , Genes Ligados a Y/genética , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/crecimiento & desarrollo , Trastornos del Desarrollo Sexual 46, XX/sangre , Trastornos del Desarrollo Sexual 46, XX/diagnóstico , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Anomalías Congénitas/sangre , Anomalías Congénitas/diagnóstico , Femenino , Humanos , Persona de Mediana Edad , Prevalencia , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
6.
Differentiation ; 103: 46-65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30236463

RESUMEN

Development of the human female reproductive tract is reviewed from the ambisexual stage to advanced development of the uterine tube, uterine corpus, uterine cervix and vagina at 22 weeks. Historically this topic has been under-represented in the literature, and for the most part is based upon hematoxylin and eosin stained sections. Recent immunohistochemical studies for PAX2 (reactive with Müllerian epithelium) and FOXA1 (reactive with urogenital sinus epithelium and its known pelvic derivatives) shed light on an age-old debate on the derivation of vaginal epithelium supporting the idea that human vaginal epithelium derives solely from urogenital sinus epithelium. Aside for the vagina, most of the female reproductive tract is derived from the Müllerian ducts, which fuse in the midline to form the uterovaginal canal, the precursor of uterine corpus and uterine cervix an important player in vaginal development as well. Epithelial and mesenchymal differentiation markers are described during human female reproductive tract development (keratins, homeobox proteins (HOXA11 and ISL1), steroid receptors (estrogen receptor alpha and progesterone receptor), transcription factors and signaling molecules (TP63 and RUNX1), which are expressed in a temporally and spatially dynamic fashion. The utility of xenografts and epithelial-mesenchymal tissue recombination studies are reviewed.


Asunto(s)
Genitales Femeninos/crecimiento & desarrollo , Conductos Paramesonéfricos/crecimiento & desarrollo , Útero/crecimiento & desarrollo , Vagina/crecimiento & desarrollo , Femenino , Genitales Femeninos/metabolismo , Proteínas de Homeodominio/genética , Humanos , Proteínas con Homeodominio LIM/genética , Receptores de Progesterona/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
7.
Differentiation ; 97: 9-22, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28918284

RESUMEN

We present a detailed review of the embryonic and fetal development of the human female reproductive tract utilizing specimens from the 5th through the 22nd gestational week. Hematoxylin and eosin (H&E) as well as immunohistochemical stains were used to study the development of the human uterine tube, endometrium, myometrium, uterine cervix and vagina. Our study revisits and updates the classical reports of Koff (1933) and Bulmer (1957) and presents new data on development of human vaginal epithelium. Koff proposed that the upper 4/5ths of the vagina is derived from Müllerian epithelium and the lower 1/5th derived from urogenital sinus epithelium, while Bulmer proposed that vaginal epithelium derives solely from urogenital sinus epithelium. These conclusions were based entirely upon H&E stained sections. A central player in human vaginal epithelial development is the solid vaginal plate, which arises from the uterovaginal canal (fused Müllerian ducts) cranially and squamous epithelium of urogenital sinus caudally. Since Müllerian and urogenital sinus epithelium cannot be unequivocally identified in H&E stained sections, we used immunostaining for PAX2 (reactive with Müllerian epithelium) and FOXA1 (reactive with urogenital sinus epithelium). By this technique, the PAX2/FOXA1 boundary was located at the extreme caudal aspect of the vaginal plate at 12 weeks. During the ensuing weeks, the PAX2/FOXA1 boundary progressively extended cranially such that by 21 weeks the entire vaginal epithelium was FOXA1-reactive and PAX2-negative. This observation supports Bulmer's proposal that human vaginal epithelium derives solely from urogenital sinus epithelium. Clearly, the development of the human vagina is far more complex than previously envisioned and appears to be distinctly different in many respects from mouse vaginal development.


Asunto(s)
Factor Nuclear 3-alfa del Hepatocito/genética , Conductos Paramesonéfricos/crecimiento & desarrollo , Factor de Transcripción PAX2/genética , Vagina/crecimiento & desarrollo , Animales , Desarrollo Embrionario/genética , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Conductos Paramesonéfricos/metabolismo , Reproducción/genética , Vagina/metabolismo
8.
Differentiation ; 97: 54-72, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29053991

RESUMEN

Human female reproductive tract development rests mostly upon hematoxilyn and eosin stained sections despite recent advances on molecular mechanisms in mouse studies. We report application of immunohistochemical methods to explore the ontogeny of epithelial and mesenchymal differentiation markers (keratins, homobox proteins, steroid receptors), transcription factors and signaling molecules (TP63 and RUNX1) during human female reproductive tract development. Keratins 6, 7, 8, 10, 14 and 19 (KRT6, KRT7, KRT8, KRT10, KRT14, KRT19) were expressed in a temporally and spatially dynamic fashion. The undifferentiated Müllerian duct and uterovaginal canal, lined by simple columnar epithelia, expressed KRT7, KRT8 and KRT19. Glandular derivatives of the Müllerian duct (uterine tube, uterine corpus and endocervix) maintained expression of these keratins, while tissues that undergo stratified squamous differentiation (exocervix and vagina) expressed KRT6, KRT14 and KRT10 during development in an age-dependent fashion. TP63 and RUNX1 were expressed prior to KRT14, as these two transcription factors are known to be upstream from KRT14 in developing Müllerian epithelium. In the vagina, KRT10, a marker of terminal differentiation, appeared after endogenous estrogens transformed the epithelium to a thick glycogenated squamous epithelium. Uroplakin, a protein unique to urothelium, was expressed only in the bladder, urethra and vaginal introitus, but not in the female reproductive tract itself. Mesenchymal differentiation was examined through immunostaining for HOXA11 (expressed in uterine mesenchyme) and ISL1 (expressed in vaginal mesenchyme). A detailed ontogeny of estrogen receptor alpha (ESR1), progesterone receptor (PGR) and the androgen receptor (AR) provides the mechanistic underpinning for the teratogenicity of estrogens, progestins and androgens on female reproductive tract development. Immunohistochemical analysis of differentiation markers and signaling molecules advance our understanding of normal development of the human female reproductive tract. These observations demonstrate remarkable similarities in mouse and human female reproductive tract development, but also highlight some key differences.


Asunto(s)
Genitales Femeninos/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Queratinas/genética , Receptores de Esteroides/genética , Reproducción/genética , Animales , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genitales Femeninos/metabolismo , Humanos , Ratones , Conductos Paramesonéfricos/crecimiento & desarrollo , Conductos Paramesonéfricos/metabolismo , Receptores Androgénicos/genética , Receptores de Progesterona , Útero/crecimiento & desarrollo , Útero/metabolismo , Vagina/crecimiento & desarrollo , Vagina/metabolismo
9.
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
10.
Differentiation ; 87(1-2): 23-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24433705

RESUMEN

Marsupials differ from eutherian mammals in their reproductive strategy of delivering a highly altricial young after a short gestation. The young, with its undeveloped organ systems completes its development post-natally, usually within a pouch. The young is dependent on milk with a composition that varies through lactation to support its growth and changing needs as it matures over a lengthy period. Gonadal differentiation occurs after birth, providing a unique opportunity to examine the effects of hormonal manipulations on its sexual differentiation of the highly accessible young. In marsupials a difference in the migration of the urinary ducts around the genital ducts from eutherian mammals results in the unique tammar reproductive tract which has three vaginae and two cervices, and two distinctly separate uteri. In the tammar wallaby, a small member of the kangaroo family, we showed that virilisation of the Wolffian duct, prostate and phallus depends on an alternate androgen pathway, which has now been shown to be important for virilisation in humans. Through hormonal manipulations over differing time periods we have achieved sex reversal of both ovaries and testes, germ cells, genital ducts, prostate and phallus. Whilst we understand many of the mechanisms behind sexual differentiation there are still many lessons to be learned from understanding how sex reversal is achieved by using a model such as the tammar wallaby. This will help guide investigations into the major questions of how and why sex determination is achieved in other species. This review discusses the control and development of the marsupial urogenital system, largely drawn from our studies in the tammar wallaby and our ability to manipulate this system to induce sex reversal.


Asunto(s)
Andrógenos/metabolismo , Marsupiales/crecimiento & desarrollo , Diferenciación Sexual/genética , Sistema Urogenital/crecimiento & desarrollo , Animales , Femenino , Gónadas/crecimiento & desarrollo , Masculino , Marsupiales/genética , Conductos Paramesonéfricos/crecimiento & desarrollo , Testículo/crecimiento & desarrollo
11.
J Reprod Med ; 58(11-12): 517-28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24568047

RESUMEN

OBJECTIVE: To determine whether the process of vitrification affects the development of the female Müllerian duct. STUDY DESIGN: We examined the difference in morphology and gene expression of the developing uteri of 30 female mice from transferred vitrified embryos and 30 female mice from transferred fresh embryos. RESULTS: By employing histology, the data showed that there were no significant differences between the 2 groups of the same age. With the use of reverse transcription polymerase chain reaction and Western blotting, the data showed that there was no significant change in the expression of Wnt genes (Wnt4, Wnt5a, Wnt7a), beta-catenin/TCF target genes, and homeobox A10 (HOXA10) gene during uterine development in the vitrified group as compared with the control group. CONCLUSION: These data suggest that vitrifying preimplantation embryos may have no effects on morphology and gene expression of the uterus of offspring.


Asunto(s)
Blastocisto/fisiología , Criopreservación/veterinaria , Útero/crecimiento & desarrollo , beta Catenina/análisis , Animales , Transferencia de Embrión/veterinaria , Femenino , Expresión Génica , Proteínas Homeobox A10 , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/genética , Ratones , Conductos Paramesonéfricos/crecimiento & desarrollo , Embarazo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Útero/metabolismo , Proteínas Wnt/análisis , Proteínas Wnt/genética , beta Catenina/genética
12.
Congenit Anom (Kyoto) ; 63(2): 30-39, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36517931

RESUMEN

Müllerian ducts give rise to the oviducts, uterus, cervix, and vagina. During female reproductive tract development in mice, the bilateral Müllerian duct epithelium grows caudally until reaching the urogenital sinus epithelium. This is followed by further caudal growth with the reduction of the urogenital sinus epithelium. Finally, the vaginal epithelium of adult mice is entirely derived from the Müllerian duct epithelium. Here, we explored the mechanisms underlying mouse vaginal development via cell proliferation, apoptosis, and lineage analyses. We found that at the late embryonic stages, apoptosis occurred at the attachment site of bilateral Müllerian duct epithelia below the cervix, resulting in bilateral lumen traffic. The Müllerian duct epithelium was enclosed by the urogenital sinus epithelium at their boundary region on embryonic day (E) 16.5, whereas the Müllerian duct epithelium encased the urogenital sinus epithelium at postnatal day (P) 0 through lateral enlargement. Lateral Müllerian duct enlargement was accompanied by focal ERK activation within the curved epithelial tips and the specific localization of mitotic nuclei on the luminal side of the Müllerian duct epithelial layer at E17.5. Descent of the Müllerian duct epithelium and shortening of the urogenital sinus epithelium occurred rapidly after birth, accompanied by cell proliferation in the Müllerian duct epithelium and its peripheral mesenchymal tissues as well as intense apoptosis in the urogenital sinus epithelium around their boundary region. Urogenital sinus epithelium was localized at the base of the vagina at P7. In conclusion, the mouse vagina develops laterally at the late embryonic stages and caudally after birth.


Asunto(s)
Vagina , Animales , Femenino , Ratones , Epitelio , Conductos Paramesonéfricos/crecimiento & desarrollo , Útero , Vagina/crecimiento & desarrollo
13.
Reprod Biol ; 21(3): 100537, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34271244

RESUMEN

The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin Motifs) enzymes are secreted metalloproteinases with major roles in development, morphogenesis, and tissue repair via the assembly and degradation of extracellular matrix (ECM). In this study, we investigated the role of ADAMTS18 in the development of the reproductive tract in female mice by phenotyping Adamts18 knockout (Adamts18-/-) mice. The results showed that Adamst18 mRNAs were abundantly expressed in vaginal epithelial cells and muscularis cells of the developing vagina. At the time of vaginal opening (5 weeks of age), about 41 % of Adamts18-/- females showed enlarged protrusions in the upper and middle parts of the vagina, reduced vaginal length, and simultaneously exhibited vaginal atresia. 6% Adamts18-/- females exhibited vaginal septum. Histological analyses revealed that the paired Mullerian ducts in ∼33 % female Adamts18-/- embryos failed to fuse at embryonic day 15.5 (E15.5) resulting in the formation of two vaginal cavities. Results of TUNEL assay and immunohistochemistry for caspase-3 showed that the number of apoptotic cells in the terminal portion of the vagina of 5-week-old Adamts18-/- females with vaginal atresia was significantly decreased. Adamts18-/- females also showed a significant decrease in serum estradiol E2 compared to age-matched Adamts18+/+ females. Results of qRT-PCR showed that the expression level of the anti-apoptosis gene Bcl-2 was significantly increased and that of the apoptosis-related gene Epha1 was decreased in the vagina of 5-week-old Adamts18-/- females. These results suggest that ADAMTS18 regulates vaginal opening through influencing the fusion of Mullerian ducts and apoptosis of vaginal cells in mice.


Asunto(s)
Proteínas ADAMTS/metabolismo , Células Epiteliales/fisiología , Conductos Paramesonéfricos/crecimiento & desarrollo , Vagina/fisiología , Proteínas ADAMTS/genética , Animales , Apoptosis , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Maduración Sexual/fisiología , Técnicas de Cultivo de Tejidos , Vagina/citología
14.
Dev Biol ; 334(2): 429-36, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19664614

RESUMEN

In developing mammalian males, conversion of the Wolffian ducts into the epididymides and vasa deferentia depends on androgen secretion by the testes, whereas in females these ducts remain in a vestigial form or regress. However, there is continuing uncertainty whether the androgen needs to be delivered locally, either by diffusion from the adjacent testis or, by secretion into the lumen of the duct, or whether circulating androgens maintain and virilize the Wolffian ducts. To resolve this uncertainty, we transplanted either day 0-2 or day 8-9 post-partum testes beneath the flank skin of three groups of neonatal (days 0-1) female tammar wallabies, where they developed and secreted physiological levels of hormones. The Wolffian ducts of all these females were retained and had formed extensive epididymides when examined at days 25, 34 and 87 after birth. In the two older groups of females, sampled after the time of prostatic bud formation, the urogenital sinus was virilized and there was extensive prostatic development similar to that of normal males of the same age, showing that androgen secretion had occurred. Virilization of the Wolffian ducts occurred during an early but short-lived window of sensitivity. This study provides the first clear evidence that under physiological conditions virilization can be mediated by circulating androgen.


Asunto(s)
Andrógenos/fisiología , Diferenciación Sexual/fisiología , Testículo/metabolismo , Sistema Urogenital/crecimiento & desarrollo , Virilismo/etiología , Conductos Mesonéfricos/crecimiento & desarrollo , Andrógenos/sangre , Andrógenos/metabolismo , Animales , Animales Recién Nacidos , Epidídimo/crecimiento & desarrollo , Femenino , Macropodidae , Masculino , Morfogénesis , Conductos Paramesonéfricos/crecimiento & desarrollo , Próstata/crecimiento & desarrollo , Tejido Subcutáneo , Testículo/trasplante , Factores de Tiempo , Trasplante Heterólogo , Virilismo/fisiopatología
15.
Reprod Toxicol ; 86: 56-61, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30940504

RESUMEN

In female mice, the Müllerian duct develops into the oviduct, uterus and vagina. The fate of epithelia is determined by factors secreted from the mesenchyme. Retinoic acid (RA) and its receptors are present in the mesenchyme of cranial Müllerian duct. RA induces Müllerian duct to uterine epithelial differentiation whereas inhibition of RA receptors induces vaginal epithelial differentiation. Thus, RA signaling in the Müllerian duct is required to promote differentiation of the mesenchyme into the future uterus. Perinatal estrogen exposure induces various abnormalities in Müllerian duct-derived organs. These include a cranial shift of the border among oviduct, uterus and vagina as well as precancerous lesions suppressed by co-treatment with RA and estrogen. Since RA synthesis enzymes and receptors are expressed both in the epithelium and stroma after birth, RA signaling may act in the epithelia to maintain adult epithelial homeostasis and to prevent irreversible lesions induced by perinatal estrogen exposure.


Asunto(s)
Conductos Paramesonéfricos/crecimiento & desarrollo , Tretinoina/fisiología , Útero/crecimiento & desarrollo , Animales , Estrógenos/metabolismo , Femenino , Humanos , Transducción de Señal
16.
Genetics ; 213(2): 529-553, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31399485

RESUMEN

Fetal mammalian testes secrete Anti-Müllerian hormone (Amh), which inhibits female reproductive tract (Müllerian duct) development. Amh also derives from mature mammalian ovarian follicles, which marks oocyte reserve and characterizes polycystic ovarian syndrome. Zebrafish (Danio rerio) lacks Müllerian ducts and the Amh receptor gene amhr2 but, curiously, retains amh To discover the roles of Amh in the absence of Müllerian ducts and the ancestral receptor gene, we made amh null alleles in zebrafish. Results showed that normal amh prevents female-biased sex ratios. Adult male amh mutants had enormous testes, half of which contained immature oocytes, demonstrating that Amh regulates male germ cell accumulation and inhibits oocyte development or survival. Mutant males formed sperm ducts and some produced a few offspring. Young female mutants laid a few fertile eggs, so they also had functional sex ducts. Older amh mutants accumulated nonvitellogenic follicles in exceedingly large but sterile ovaries, showing that Amh helps control ovarian follicle maturation and proliferation. RNA-sequencing data partitioned juveniles at 21 days postfertilization (dpf) into two groups that each contained mutant and wild-type fish. Group21-1 upregulated ovary genes compared to Group21-2, which were likely developing as males. By 35 dpf, transcriptomes distinguished males from females and, within each sex, mutants from wild types. In adult mutants, ovaries greatly underexpressed granulosa and theca genes, and testes underexpressed Leydig cell genes. These results show that ancestral Amh functions included development of the gonadal soma in ovaries and testes and regulation of gamete proliferation and maturation. A major gap in our understanding is the identity of the gene encoding a zebrafish Amh receptor; we show here that the loss of amhr2 is associated with the breakpoint of a chromosome rearrangement shared among cyprinid fishes.


Asunto(s)
Hormona Antimülleriana/genética , Genitales Femeninos/crecimiento & desarrollo , Procesos de Determinación del Sexo , Pez Cebra/genética , Animales , Femenino , Gónadas/crecimiento & desarrollo , Conductos Paramesonéfricos/crecimiento & desarrollo , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , RNA-Seq , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Pez Cebra/crecimiento & desarrollo
17.
Methods Mol Biol ; 1965: 173-186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31069675

RESUMEN

The reproductive cycle encompasses processes such as sex organ differentiation and development in the early life stages and maturation of the gametes in the adult organism. During the early life stages, critical developmental programming of the endocrine and reproductive systems occurs, and exposure to chemicals during these critical developmental windows can result in impaired reproductive function later in life. It is therefore important to evaluate long-term consequences of early life stage exposure to endocrine-disrupting chemicals. The African clawed frog Xenopus tropicalis has several characteristics that facilitate studies of developmental and reproductive toxicity. Here I present a X. tropicalis life cycle test protocol including study design, exposure regimes, and endpoints for chemical disruption of sex differentiation, gonadal and Müllerian duct development, the thyroxin-regulated metamorphosis, estrogen synthesis (activity of the CYP19 aromatase enzyme), spermatogenesis, oogenesis, puberty and fertility.


Asunto(s)
Gametogénesis/efectos de los fármacos , Conductos Paramesonéfricos/crecimiento & desarrollo , Xenopus/crecimiento & desarrollo , Animales , Embrión no Mamífero/efectos de los fármacos , Femenino , Estadios del Ciclo de Vida/efectos de los fármacos , Masculino , Metamorfosis Biológica/efectos de los fármacos , Modelos Animales , Conductos Paramesonéfricos/efectos de los fármacos , Diferenciación Sexual , Tiroxina/metabolismo , Xenopus/metabolismo
18.
J Pediatr Surg ; 52(10): 1656-1660, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28599968

RESUMEN

BACKGROUND/AIM: Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance (MIS), is glycoprotein hormone secreted by the fetal Sertoli cells to regulate regression of the Müllerian ducts, the anlagen of the uterus, fallopian tubes, and upper vagina. After its existence was predicted in 1946 and its isolation and purification in the 1970's, a huge amount of information has been gathered on its molecular biology and function in the last 30-40years. Once thought to be a locally acting factor in the male fetus during sexual differentiation, it is now recognized as an endocrine hormone present in both sexes and with functions throughout life. One of the remaining controversies is the possible role of AMH during fetal testicular descent. In the human with aberrant AMH function, the boy has cryptorchidism with persistent Müllerian duct syndrome (PMDS), where the testes are often intraabdominal and on an abnormally long gubernacular cord. By contrast, in rodent models knockout of the AMH gene does not cause cryptorchidism. METHODS/RESULTS: In this review we examined the evidence in the literature for and against a role for AMH in testicular descent and considered the implications of the different anatomy of the gubernacular cord in rodents versus children. CONCLUSION: We conclude that AMH may have a role in shortening the gubernacular cord in humans which is concealed in rodent models by differences in anatomy of the gubernacular cord in rodents. The controversy could be resolved by re-examination of the gubernacular cord in boys with PMDS and mice with AMHKO. TYPE OF STUDY: Review. LEVEL OF EVIDENCE: V.


Asunto(s)
Hormona Antimülleriana/metabolismo , Criptorquidismo/metabolismo , Inhibidores de Crecimiento/metabolismo , Conductos Paramesonéfricos/metabolismo , Animales , Trastorno del Desarrollo Sexual 46,XY , Trompas Uterinas/metabolismo , Femenino , Humanos , Masculino , Ratones , Conductos Paramesonéfricos/crecimiento & desarrollo , Células de Sertoli/metabolismo
19.
Sci Rep ; 6: 23037, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26964900

RESUMEN

Congenital reproductive tract anomalies could impair fertility. Female and male reproductive tracts are developed from Müllerian ducts and Wolffian ducts, respectively, involving initiation, elongation and differentiation. Genetic basis solely for distal reproductive tract development is largely unknown. Lhfpl2 (lipoma HMGIC fusion partner-like 2) encodes a tetra-transmembrane protein with unknown functions. It is expressed in follicle cells of ovary and epithelial cells of reproductive tracts. A spontaneous point mutation of Lhfpl2 (LHFPL2(G102E)) leads to infertility in 100% female mice, which have normal ovarian development, ovulation, uterine development, and uterine response to exogenous estrogen stimulation, but abnormal upper longitudinal vaginal septum and lower vaginal agenesis. Infertility is also observed in ~70% mutant males, which have normal mating behavior and sperm counts, but abnormal distal vas deferens convolution resulting in complete and incomplete blockage of reproductive tract in infertile and fertile males, respectively. On embryonic day 15.5, mutant Müllerian ducts and Wolffian ducts have elongated but their duct tips are enlarged and fail to merge with the urogenital sinus. These findings provide a novel function of LHFPL2 and a novel genetic basis for distal reproductive tract development; they also emphasize the importance of an additional merging phase for proper reproductive tract development.


Asunto(s)
Genitales/crecimiento & desarrollo , Genitales/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Infertilidad Femenina/genética , Reproducción/genética , Animales , Femenino , Pérdida Auditiva Sensorineural/genética , Infertilidad Femenina/patología , Masculino , Ratones , Conductos Paramesonéfricos/crecimiento & desarrollo , Conductos Paramesonéfricos/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Mutación Puntual , Diferenciación Sexual/genética , Sistema Urogenital/crecimiento & desarrollo , Sistema Urogenital/metabolismo , Sistema Urogenital/patología , Conductos Mesonéfricos/crecimiento & desarrollo , Conductos Mesonéfricos/metabolismo
20.
J Mol Endocrinol ; 8(1): 73-7, 1992 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1543536

RESUMEN

To determine whether mammalian Müllerian-inhibiting substance (MIS) is active in birds, Müllerian ducts from 7- to 8-day-old male or female chick embryos were cultured in the presence of human recombinant MIS at concentrations between 2.5 and 12.5 micrograms/ml. None of 20 ducts regressed at any concentration. In contrast, at concentrations of 2.5-5 micrograms/ml, all 12 Müllerian ducts from 13-day-old male mouse embryos and 13 out of 14 female ducts were inhibited to varying degrees. It is concluded that avian Müllerian ducts are unresponsive to mammalian MIS. There may be a difference in structure between the MIS of birds and mammals, or the signal-transduction system may be different.


Asunto(s)
Glicoproteínas , Inhibidores de Crecimiento/farmacología , Conductos Paramesonéfricos/efectos de los fármacos , Hormonas Testiculares/farmacología , Animales , Hormona Antimülleriana , Embrión de Pollo , Femenino , Humanos , Masculino , Ratones , Conductos Paramesonéfricos/crecimiento & desarrollo , Técnicas de Cultivo de Órganos , Proteínas Recombinantes/farmacología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA