Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
J Pediatr Endocrinol Metab ; 34(10): 1247-1255, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34265880

RESUMEN

OBJECTIVES: Antimullerian hormone (AMH) causes regression of the mullerian ducts in the male fetus. The appendix testis (AT) is a vestigial remnant of mullerian duct origin, containing both androgen (AR) and estrogen (ER) receptors. The role of both AMH and AT in testicular descent is yet to be studied. We investigated the possible association of AMH with AT size, the AR and ER, and their expression in the AT, in congenital cryptorchidism. METHODS: A total of 26 patients with congenital unilateral cryptorchidism and 26 controls with orthotopic testes were investigated, and 21 ATs were identified in each group. AMH and insulin-like three hormone (INSL3) concentrations were measured with spectrophotometry. AR and ER receptor expression was assessed with immunohistochemistry using monoclonal antibodies R441 for AR and MAB463 for ER. For the estimation of receptor expression, the Allred Score method was used. RESULTS: AMH concentrations did not present significant differences between patients with congenital cryptorchidism and the controls. Also, no correlation was found between AMH, INSL3, and AT length. Allred scores did not present significant differences. However, expression percentiles and intensity for both receptors presented significant differences. Three children with cryptorchidism and the highest AMH levels also had the highest estrogen receptor scores in the AT. CONCLUSIONS: No association was found between AMH and the studied major parameters. However, higher AMH concentrations, in combination with higher estrogen receptor scores in the AT, may play a role in cryptorchidism in some children. Larger population samples are needed to verify this observation.


Asunto(s)
Hormona Antimülleriana/sangre , Criptorquidismo/patología , Genitales Masculinos/patología , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Preescolar , Estudios de Cohortes , Criptorquidismo/sangre , Criptorquidismo/genética , Expresión Génica , Genitales Masculinos/anomalías , Genitales Masculinos/embriología , Grecia , Humanos , Lactante , Insulina/sangre , Masculino , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/metabolismo , Conductos Paramesonéfricos/patología , Tamaño de los Órganos , Proteínas , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Testículo/anomalías , Testículo/patología
2.
J Endocrinol ; 247(1): R27-R44, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32755997

RESUMEN

The spotted hyaena (Crocuta crocuta) is a unique species, even amongst the Hyaenidae. Extreme clitoral development in female spotted hyaenas challenges aspects of the accepted framework of sexual differentiation and reproductive function. They lack a vulva and instead urinate, copulate and give birth through a single, long urogenital canal that traverses a clitoris superficially resembling a penis. Recent and historical evidence is reviewed to describe our changing understanding of the biology of this species. Expanding upon observations from hyaenas in nature, much has been learned from studies utilising the captive colony at the University of California, Berkeley. The steroid environment of pregnancy is shaped by placental androgen and oestrogen secretion and a late gestational increase in sex hormone binding globulin, the regulated expression and steroid-binding characteristics of which are unique within the Hyaenidae. While initial external genital development is largely free of androgenic influence, the increase in testosterone concentrations in late gestation influences foetal development. Specifically, anti-androgen (AA) treatment of pregnant females reduced the developmental influence of androgens on their foetuses, resulting in reduced androstenedione concentrations in young females and easier birth through a 'feminised' clitoris, but precluded intromission and mating by 'feminised' male offspring, and altered social interactions. Insight into the costs and benefits of androgen exposure on spotted hyaena reproductive development, endocrinology and behaviour emphasises the delicate balance that sustains reproductive success, forces a re-evaluation of how we define masculine vs feminine sexual characteristics, and motivates reflection about the representative value of model species.


Asunto(s)
Genitales Femeninos , Genitales Masculinos , Hormonas Esteroides Gonadales/fisiología , Hyaenidae , Reproducción/fisiología , Diferenciación Sexual/fisiología , Andrógenos/fisiología , Animales , Estrógenos/fisiología , Femenino , Genitales Femeninos/anatomía & histología , Genitales Femeninos/embriología , Genitales Femeninos/crecimiento & desarrollo , Genitales Masculinos/anatomía & histología , Genitales Masculinos/embriología , Genitales Masculinos/crecimiento & desarrollo , Hyaenidae/anatomía & histología , Hyaenidae/embriología , Hyaenidae/fisiología , Masculino , Embarazo , Globulina de Unión a Hormona Sexual/fisiología , Conducta Sexual Animal/fisiología
3.
Nat Rev Urol ; 15(6): 358-368, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29670181

RESUMEN

The biology of masculinization is fundamentally important for understanding the embryonic developmental processes that are involved in the development of the male reproductive tract, external genitalia, and also the tumorigenesis of prostate cancer. The molecular mechanisms of masculinization are of interest to many researchers and clinicians involved in varied fields, including molecular developmental biology, cancer research, endocrinology, and urology. Androgen signalling is mediated by the nuclear androgen receptor, which has fundamental roles in masculinization during development. Various modes of androgen signalling, including 5α-dihydrotestosterone-induced regulation of mesenchymal cell proliferation, have been observed in masculinization. Such regulation is essential for regulating urogenital tissue development, including external genitalia development. Androgen-induced genes, such as MAFB, which belongs to the activator protein 1 (AP-1) superfamily of genes, have essential roles in male urethral formation, and disruption of its signalling can interfere with urethral formation, which often results in hypospadias. Another AP-1 superfamily gene, ATF3, could be responsible for some instances of hypospadias in humans. These androgen-dependent signals and downstream events are crucial for not only developmental processes but also processes of diseases such as hypospadias and prostate cancer.


Asunto(s)
Andrógenos/metabolismo , Genitales Masculinos/embriología , Receptores Androgénicos/metabolismo , Diferenciación Sexual/fisiología , Biomarcadores/metabolismo , Genitales Masculinos/anomalías , Genitales Masculinos/metabolismo , Humanos , Hipospadias/embriología , Hipospadias/metabolismo , Masculino , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/metabolismo , Transducción de Señal
4.
Ann Chir Plast Esthet ; 61(5): 439-449, 2016 Oct.
Artículo en Francés | MEDLINE | ID: mdl-27177771

RESUMEN

Hypospadias is a hypoplasia of the tissues forming the ventral side of the penis responsible of an ectopic meatus of the urethra. This congenital anomaly results in a fusion defect of the two epithelial surfaces of the urethral groove between the 11th and the 18th weeks of development. The earlier this process arrests, the more the form is proximal and severe. This is the second genital malformation in boys with 1 case per 250 male births. Its origin is often multifactorial (genetic, endocrine, placental and environmental). Three anatomical forms exist: proximal, middle and distal (the most common). Additional exams (endocrine, genetic and morphological) are realized early, before surgery, in case of severe hypospadias, familial, associated with cryptorchidism, bifid scrotum, micropenis and/or skeletal, kidney, and/or heart abnormalities. It clarifies pubertal prognosis. The surgical management is made between 6months and 12months: it limits the functional and aesthetic impact of this malformation. Many surgical techniques are described. They all have in common the three operating time: penile straightening, urethroplasty, reconstruction of the ventral side of penis. They are based on direct sutures, local flaps pedicled, and grafts (skin or mucosa). The rate of postoperative complications is between 6 and 30 %. The two main complications are fistulae and stenoses. The psychological follow-up of these children is necessary to adulthood. Surgery of hypospadias remains a delicate surgery and must be performed by experienced surgeons.


Asunto(s)
Hipospadias/cirugía , Procedimientos de Cirugía Plástica/métodos , Procedimientos Quirúrgicos Urológicos Masculinos/métodos , Genitales Masculinos/embriología , Humanos , Masculino , Factores de Riesgo
5.
J Urol ; 194(6): 1728-36, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26162297

RESUMEN

PURPOSE: Genetic variants in diacylglycerol kinase κ (DGKK) have been strongly associated with risk of hypospadias. We investigated the expression pattern of Dgkk during development of mouse external genitalia to better understand its function and mechanism in the etiology of hypospadias. MATERIALS AND METHODS: We performed Dgkk expression analysis via indirect immunofluorescence in histological sections of CD-1 mouse embryonic and postnatal male, female and diethylsilbestrol treated external genitalia. Histological findings were supplemented with DGKK expression analysis using quantitative real-time polymerase chain reaction assays. RESULTS: In mouse external genitalia Dgkk was expressed in the membrane and cytoplasm of differentiating squamous epithelial cells of urethral plate/groove and epidermis but not in the undifferentiated epithelial cells of preputial lamina or basal layer of urethral groove epithelium. CD-1 gestation day 18 male mouse genital tubercle treated with oil or diethylstilbestrol showed similar patterns of Dgkk expression despite many morphological differences, including formation of preputial cleft observed in diethylsilbestrol treated mice. CONCLUSIONS: Dgkk appears to be a marker or mediator of squamous cell differentiation during development of mouse external genitalia. However, no association exists between Dgkk expression and formation of preputial cleft in the genital tubercle of diethylsilbestrol treated mice, suggesting that these 2 events may follow independent pathways in mice. Further studies are needed to elucidate the role of DGKK in hypospadias.


Asunto(s)
Diacilglicerol Quinasa/genética , Variación Genética/genética , Genitales Masculinos/embriología , Hipospadias/embriología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Dietilestilbestrol/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Variación Genética/efectos de los fármacos , Genitales Masculinos/efectos de los fármacos , Edad Gestacional , Hipospadias/genética , Masculino , Ratones , Uretra/embriología
6.
Proc Natl Acad Sci U S A ; 111(46): 16407-12, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25362053

RESUMEN

Masculinization of external genitalia is an essential process in the formation of the male reproductive system. Prominent characteristics of this masculinization are the organ size and the sexual differentiation of the urethra. Although androgen is a pivotal inducer of the masculinization, the regulatory mechanism under the control of androgen is still unknown. Here, we address this longstanding question about how androgen induces masculinization of the embryonic external genitalia through the identification of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb) gene. Mafb is expressed prominently in the mesenchyme of male genital tubercle (GT), the anlage of external genitalia. MAFB expression is rarely detected in the mesenchyme of female GTs. However, exposure to exogenous androgen induces its mesenchymal expression in female GTs. Furthermore, MAFB expression is prominently down-regulated in male GTs of androgen receptor (Ar) KO mice, indicating that AR signaling is necessary for its expression. It is revealed that Mafb KO male GTs exhibit defective embryonic urethral formation, giving insight into the common human congenital anomaly hypospadias. However, the size of Mafb KO male GTs is similar with that of wild-type males. Moreover, androgen treatment fails to induce urethral masculinization of the GTs in Mafb KO mice. The current results provide evidence that Mafb is an androgen-inducible, sexually dimorphic regulator of embryonic urethral masculinization.


Asunto(s)
Genitales Masculinos/embriología , Factor de Transcripción MafB/fisiología , Mesodermo/metabolismo , Caracteres Sexuales , Diferenciación Sexual/fisiología , Uretra/embriología , Andrógenos/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Genitales Masculinos/metabolismo , Hipospadias/embriología , Hipospadias/genética , Factor de Transcripción MafB/biosíntesis , Factor de Transcripción MafB/deficiencia , Factor de Transcripción MafB/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Androgénicos/deficiencia , Receptores Androgénicos/fisiología , Uretra/anomalías , Uretra/metabolismo
7.
Vitam Horm ; 94: 193-210, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24388191

RESUMEN

Methoxychlor is an organochlorine pesticide having a weak estrogenicity, which is estimated to be approximately 1000- to 14,000-fold less potent to a natural ligand, 17ß-estradiol. However, its active metabolite, hydroxyphenyltrichloroethane, has much more potent estrogenic activity and probably acts in the target organs of animals exposed to methoxychlor at least 100 times stronger than the parent compound. A variety of in vivo reproductive toxicity studies have shown that treatment with methoxychlor exerts typical endocrine-disrupting effects manifest as estrogenic effects, such as formation of cystic ovaries resulting in ovulation failures, uterine hypertrophy, hormonal imbalances, atrophy of male sexual organs, and deteriorations of sperm production in rats and/or mice, through which it causes serious reproductive damages in both sexes of animals at sufficient dose levels. However, methoxychlor is not teratogenic. The no-observed-adverse-effect level of methoxychlor among reliable experimental animal studies in terms of the reproductive toxicity is 10 ppm (equivalent to 0.600 mg/kg/day) in a two-generation reproduction toxicity study.


Asunto(s)
Estrógenos no Esteroides/toxicidad , Desarrollo Fetal/efectos de los fármacos , Genitales Femeninos/efectos de los fármacos , Genitales Masculinos/efectos de los fármacos , Insecticidas/toxicidad , Metoxicloro/toxicidad , Desarrollo Sexual/efectos de los fármacos , Animales , Biotransformación , Disruptores Endocrinos/farmacocinética , Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/farmacocinética , Contaminantes Ambientales/toxicidad , Estrógenos no Esteroides/metabolismo , Estrógenos no Esteroides/farmacocinética , Femenino , Genitales Femeninos/embriología , Genitales Femeninos/crecimiento & desarrollo , Genitales Femeninos/patología , Genitales Masculinos/embriología , Genitales Masculinos/crecimiento & desarrollo , Genitales Masculinos/patología , Humanos , Infertilidad Femenina/inducido químicamente , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Infertilidad Masculina/inducido químicamente , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Insecticidas/farmacocinética , Masculino , Metoxicloro/farmacocinética , Fenoles/metabolismo , Fenoles/toxicidad , Embarazo
8.
Asian J Androl ; 16(1): 50-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24369133

RESUMEN

During the past few decades, scientific evidence has been accumulated concerning the possible adverse effects of the exposure to environmental chemicals on the well-being of wildlife and human populations. One large and growing group of such compounds of anthropogenic or natural origin is referred to as endocrine-disrupting chemicals (EDCs), due to their deleterious action on the endocrine system. This concern was first focused on the control of reproductive function particularly in males, but has later been expanded to include all possible endocrine functions. The present review describes the underlying physiology behind the cascade of developmental events that occur during sexual differentiation of males and the specific role of androgen in the masculinization process and proper organogenesis of the external male genitalia. The impact of the genetic background, environmental exposures and lifestyle factors in the etiology of hypospadias, cryptorchidism and testicular cancer are reviewed and the possible role of EDCs in the development of these reproductive disorders is discussed critically. Finally, the possible direct and programming effects of exposures in utero to widely use therapeutic compounds, environmental estrogens and other chemicals on the incidence of reproductive abnormalities and poor semen quality in humans are also highlighted.


Asunto(s)
Trastornos del Desarrollo Sexual/inducido químicamente , Disruptores Endocrinos/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Andrógenos/fisiología , Animales , Criptorquidismo/etiología , Criptorquidismo/genética , Sistema Endocrino/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Femenino , Genitales Masculinos/embriología , Humanos , Hipospadias/etiología , Hipospadias/genética , Estilo de Vida , Masculino , Ratones , Embarazo , Ratas , Análisis de Semen , Células de Sertoli/efectos de los fármacos , Diferenciación Sexual/efectos de los fármacos , Neoplasias Testiculares/inducido químicamente , Neoplasias Testiculares/genética , Factores de Transcripción/fisiología
9.
Psychoneuroendocrinology ; 38(9): 1618-29, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23399049

RESUMEN

Depression during pregnancy and postpartum is a significant health problem and affects up to 20% of women. While selective serotonin reuptake inhibitor (SSRI) medications are the drug of choice for treatment of maternal depression, the combined effect of maternal depression and perinatal SSRI exposure on offspring development is poorly investigated. Our aim was to determine the role of exposure to fluoxetine during development on sexual behavior and sexually dimorphic brain structures in male offspring using a rodent model of maternal adversity. Sprague-Dawley rat dams were stressed during gestation and were chronically treated throughout lactation with either fluoxetine or vehicle beginning on postnatal day 1. Four groups of offspring were used: (1) Control+Vehicle, (2) Control+Fluoxetine, (3) Prenatal Stress+Vehicle, and (4) Prenatal Stress+Fluoxetine. We show here that developmental fluoxetine treatment decreases the anogenital distance in juvenile male offspring. In adult male offspring, maternal fluoxetine treatment results in a decrease in the number of intromissions, a longer latency to the first intromission, and a longer latency to the first ejaculation. Furthermore, developmental fluoxetine and/or prenatal stress decrease the area of the sexually dimorphic nucleus of the preoptic area (SDN-POA). Prenatal stress, but not exposure to developmental fluoxetine, decreases the number of tyrosine hydroxylase (TH)-positive cells in anteroventral periventricular nucleus (AVPv) and the volume of the posterior bed nucleus of the stria terminalis (pBST) in male offspring. These results provide important evidence for the long-term impact of maternal adversity and maternal fluoxetine use on the development of primary endocrinology systems in juvenile and adult male offspring.


Asunto(s)
Encéfalo/embriología , Fluoxetina/toxicidad , Complicaciones del Embarazo/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Diferenciación Sexual/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Canal Anal/embriología , Animales , Encéfalo/efectos de los fármacos , Eyaculación/fisiología , Estradiol/sangre , Femenino , Fluoxetina/farmacología , Genitales Masculinos/embriología , Masculino , Núcleos Talámicos de la Línea Media/química , Núcleos Talámicos de la Línea Media/embriología , Proteínas del Tejido Nervioso/análisis , Tamaño de los Órganos , Embarazo , Complicaciones del Embarazo/fisiopatología , Complicaciones del Embarazo/psicología , Área Preóptica/embriología , Ratas , Ratas Sprague-Dawley , Núcleos Septales/química , Núcleos Septales/embriología , Núcleos Septales/ultraestructura , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Estrés Psicológico/fisiopatología , Testosterona/sangre , Tirosina 3-Monooxigenasa/análisis
10.
PLoS One ; 8(2): e55587, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23390542

RESUMEN

Separating digestive and urinary outlets is a critical step during mammalian embryogenesis. However, the natural history of these structures is poorly studied, and little is known about their embryonic origin. Here, we show that peri-cloacal mesenchymal (PCM) progenitors are the major source of these structures. Surprisingly, PCM progenitors also contribute to perineum, a structural barrier separating the urinary and digestive tracts, suggesting a potential role of PCM progenitors in establishing independent urinary and digestive outlets. We demonstrate that Six1 and Six2 are complementarily but asymmetrically expressed in the PCM progenitors. Deletion of these genes results in decreased cell survival and proliferation, and consequently in agenesis of the perineum and severe hypoplasia of the genital tubercle. Together, these findings suggest that PCM progenitors are the unexpected source of perineum and genital tubercle, and establish a basic framework for investigating normal and abnormal development of anorectal and genitourinary structures.


Asunto(s)
Cloaca/embriología , Tracto Gastrointestinal/embriología , Genitales Femeninos/embriología , Genitales Masculinos/embriología , Proteínas de Homeodominio/genética , Perineo/embriología , Factores de Transcripción/genética , Sistema Urinario/embriología , Animales , Tipificación del Cuerpo/genética , Embrión de Mamíferos , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Transducción de Señal , Factores de Transcripción/metabolismo
11.
Mol Cell Endocrinol ; 358(1): 53-62, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22406838

RESUMEN

During embryogenesis, the development of the male genital is dependent on androgens. Their actions are mediated by the androgen receptor (AR), which functions as a transcription factor. To identify AR coregulators that support AR action during the critical time window of androgen-dependent development in the genital tubercle of male mice, we performed yeast two-hybrid screenings with cDNA libraries of genital tubercles from male mouse embryos using human AR as bait. RWD domain containing 1 (RWDD1) was identified as an AR-interacting protein from three independent libraries of the embryonic days E15, E16 and E17. The interaction between the AR and RWDD1 was confirmed in vitro and in vivo and the ligand binding domain of the AR was shown to be sufficient to mediate the interaction. RWDD1 enhanced AR-dependent transactivation in reporter assays with promoters of different complexity and in different cell lines. These results suggest that RWDD1 functions as a coactivator of androgen-dependent transcription.


Asunto(s)
Genitales Masculinos/embriología , Proteínas/metabolismo , Receptores Androgénicos/metabolismo , Activación Transcripcional , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Regulación del Desarrollo de la Expresión Génica , Genitales Masculinos/metabolismo , Células HeLa , Humanos , Masculino , Ratones , Proteínas/genética , Receptores Androgénicos/genética
12.
Int J Androl ; 35(3): 236-44, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21696396

RESUMEN

Phthalate esters, commonly used as plasticizers, show anti-androgenic activity and cause male reproductive malformation in experimental animals. However, the effects of prenatal exposure to phthalate esters in humans have not been extensively studied. The purpose of this study was to examine the relationship between prenatal exposure to phthalate esters and the anogenital distance (AGD) as a reproductive endpoint in human male newborns. Spot urine samples were collected from 111 Japanese pregnant women after obtaining their informed consent. Seven urinary phthalate ester metabolites were determined by high performance liquid chromatography-tandem mass spectrometry. Urinary isoflavones concentrations were measured as possible covariates because their oestrogenicities and high exposure levels among Japanese have the potential to affect male genital development. Birth outcomes and AGD, the distance from the centre of the anus to external genitalia, were measured for their male newborns. In a multiple regression model, the log-transformed mono-2-ethylhexyl phthalate concentration (specific gravity-corrected) was negatively significant, and maternal smoking status was positively significant, in explaining anogenital index (AGI) when potential covariates were controlled for. Urinary isoflavones did not significantly contribute to AGI in any models. Our results suggest that prenatal exposure to di(2-ethylhexyl) phthalate affects reproductive development in human males.


Asunto(s)
Ácidos Ftálicos/orina , Pueblo Asiatico , Dietilhexil Ftalato/análogos & derivados , Contaminantes Ambientales/farmacología , Equol/orina , Ésteres/farmacología , Femenino , Genitales Masculinos/efectos de los fármacos , Genitales Masculinos/embriología , Humanos , Recién Nacido , Isoflavonas/orina , Masculino , Ácidos Ftálicos/farmacología , Plastificantes/farmacología , Embarazo/orina , Efectos Tardíos de la Exposición Prenatal , Análisis de Regresión , Fumar/epidemiología
13.
J Pediatr Surg ; 46(9): 1804-12, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21929994

RESUMEN

BACKGROUND: Gubernacular development and testicular descent have been studied most extensively in rat models, but new transgenic mouse models require a deep understanding of normal mouse development so that results can be extrapolated to the human. We aimed to compare gubernacular anatomy during development in the mouse with that of the rat. METHODS: Time-mated mice (C57BL/6) and Sprague-Dawley rats were used to collect male fetuses at embryonic (E) days E13, E14, E15, E17, E18, and E19 and neonates at postnatal (P) days P0 and P2. Fetuses and newborn were processed for serial sections (sagittal, transverse, and coronal) and stained with hematoxylin and eosin, muscle markers (embryonic myosin, desmin), a neuronal marker (Tuj1), a mitotic marker (Ki67), and keratin marker to label epithelium. RESULTS: Early development of cremaster in the mouse was related to transversus abdominis muscle, but not internal oblique muscle (as in rats), and forms a monolaminar cremaster layer. There is close association between the regressing inguinal mammary bud and the gubernaculum in the mouse at E13. The peritoneal surface of the processus vaginalis (PV) covering the gubernaculum and epididymis was morphologically distinct from the remaining parietal peritoneum throughout development. CONCLUSIONS: Gubernacular development in mouse is similar to that in the rat except for certain structures, such as cremaster muscle. The PV seems to be derived from the surface of the urogenital ridge, separate from the remaining parietal peritoneum. This study suggests that the PV has evolved to aid testicular descent in this species, rather than a nondescript diverticulum of parietal peritoneum.


Asunto(s)
Genitales Masculinos/embriología , Ratones/embriología , Ratas/embriología , Animales , Masculino , Ratones Endogámicos C57BL , Músculo Liso/embriología , Ratas Sprague-Dawley , Testículo/embriología
14.
Endocrinology ; 152(7): 2894-903, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21586556

RESUMEN

During embryogenesis, sexually dimorphic organogenesis is achieved by hormones produced in the gonad. The external genitalia develop from a single primordium, the genital tubercle, and their masculinization processes depend on the androgen signaling. In addition to such hormonal signaling, the involvement of nongonadal and locally produced masculinization factors has been unclear. To elucidate the mechanisms of the sexually dimorphic development of the external genitalia, series of conditional mutant mouse analyses were performed using several mutant alleles, particularly focusing on the role of hedgehog signaling pathway in this manuscript. We demonstrate that hedgehog pathway is indispensable for the establishment of male external genitalia characteristics. Sonic hedgehog is expressed in the urethral plate epithelium, and its signal is mediated through glioblastoma 2 (Gli2) in the mesenchyme. The expression level of the sexually dimorphic genes is decreased in the glioblastoma 2 mutant embryos, suggesting that hedgehog signal is likely to facilitate the masculinization processes by affecting the androgen responsiveness. In addition, a conditional mutation of Sonic hedgehog at the sexual differentiation stage leads to abnormal male external genitalia development. The current study identified hedgehog signaling pathway as a key factor not only for initial development but also for sexually dimorphic development of the external genitalia in coordination with androgen signaling.


Asunto(s)
Genitales Masculinos/embriología , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Procesos de Determinación del Sexo , Transducción de Señal , Andrógenos/farmacología , Animales , Epitelio/efectos de los fármacos , Epitelio/embriología , Epitelio/metabolismo , Femenino , Silenciador del Gen , Genitales Femeninos/efectos de los fármacos , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Genitales Masculinos/efectos de los fármacos , Genitales Masculinos/metabolismo , Proteínas Hedgehog/genética , Hipospadias/inducido químicamente , Hipospadias/embriología , Hipospadias/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Propionato de Testosterona/farmacología , Uretra/efectos de los fármacos , Uretra/embriología , Uretra/metabolismo , Proteína Gli2 con Dedos de Zinc
15.
J Pediatr Surg ; 46(2): 387-92, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21292093

RESUMEN

BACKGROUND: The gubernaculum is central to testicular descent, with recent evidence suggesting that it elongates to the scrotum like a limb bud. Homeobox (Hox) genes involved in limb bud outgrowth are expressed within the gubernaculum. Mice with homozygous Hoxa11 gene deletions have bilateral cryptorchidism. This study investigated the precise anatomical effects of Hoxa11 mutation on the mouse gubernaculum. METHODS: The pelvises of postnatal mice (n = 46; days 1-10) with Hoxa11 knockout (n = 19), heterozygotes (n = 11), and wild-type (n = 16) mice were serially sectioned and stained with hematoxylin and eosin. Immunohistochemistry was performed for the presence of desmin. RESULTS: Hoxa11 mutant mice had intraabdominal cryptorchid testes and highly convoluted vas deferentia. The gubernacular bulbs were abnormal, with no "outgrowth" and persistence of the prenatal "swelling reaction." Desmin immunostaining revealed the lack of undifferentiated mesenchymal cells usually seen as a "swirl" within the bulb and decreased formation of cremaster muscle. CONCLUSIONS: Hoxa11 may be involved in forming the growth center seen as the "swirl" of mesenchyme within the gubernacular bulb, consistent with these cells being required for gubernacular elongation during testicular descent. Hoxa11 mutations may well contribute to failure of gubernacular migration in boys with cryptorchidism.


Asunto(s)
Proteínas de Homeodominio/fisiología , Testículo/anatomía & histología , Testículo/embriología , Andrógenos/genética , Andrógenos/fisiología , Animales , Criptorquidismo/embriología , Criptorquidismo/genética , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/genética , Genitales Masculinos/anatomía & histología , Genitales Masculinos/embriología , Proteínas de Homeodominio/genética , Humanos , Masculino , Mesodermo/embriología , Mesodermo/crecimiento & desarrollo , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación/genética , Testículo/crecimiento & desarrollo
16.
Int J Androl ; 34(4 Pt 2): e49-58, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21091719

RESUMEN

Second- to fourth-digit length ratio, 2D:4D, is a marker of testosterone level during foetal life that was found associated with sperm concentration or testosterone levels in some studies, but not in others, a difference possibly related to the way the ratio is assessed. In this study, 2D:4D was assessed in 122 men partners of pregnant women and in 71 testicular cancer patients using a new method based on direct measurements of finger lengths. In addition, we investigated the association between 2D:4D, birth weight, testicular volume, semen quality and time to pregnancy. A validation study of the method demonstrated high reliability and reproducibility. Neither digit lengths nor 2D:4D significantly differed in both groups of men. We found a significant negative association between 2D:4D and birth weight in testicular cancer patients. In fertile men, 2D:4D was associated with testicular volume (r=-0.36, p<0.001), total sperm number (r=-0.18, p=0.04) and time to pregnancy (r=0.24, p<0.02). In addition, participants with a history of epididymal cyst had a significantly higher 2D:4D than those without cysts. In conclusion, all significant findings indicate that the human male reproductive function is negatively related to 2D:4D. However, 2D:4D for testicular cancer patients does not point to a hormonal imbalance during foetal life as the common cause for developing germ-cell cancer. Such results obtained, thanks to an easy, direct and reliable method for measuring finger lengths, suggest the usefulness of this new tool in fertility studies as well as for studying men with developmental disorders of the reproductive tract.


Asunto(s)
Dedos/anatomía & histología , Genitales Masculinos/química , Genitales Masculinos/embriología , Análisis de Semen , Neoplasias Testiculares/química , Neoplasias Testiculares/patología , Testosterona/análisis , Biomarcadores , Fertilidad/fisiología , Fármacos para la Fertilidad Masculina , Genitales Masculinos/metabolismo , Humanos , Masculino , Salud Reproductiva , Semen/citología , Neoplasias Testiculares/metabolismo , Testosterona/biosíntesis
17.
Dev Biol ; 344(2): 1071-87, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20510229

RESUMEN

Here we describe the first detailed catalog of gene expression in the developing lower urinary tract (LUT), including epithelial and mesenchymal portions of the developing bladder, urogenital sinus, urethra, and genital tubercle (GT) at E13 and E14. Top compartment-specific genes implicated by the microarray data were validated using whole-mount in situ hybridization (ISH) over the entire LUT. To demonstrate the potential of this resource to implicate developmentally critical features, we focused on gene expression patterns and pathways in the sexually indeterminate, androgen-independent GT. GT expression patterns reinforced the proposed similarities between development of GT, limb, and craniofacial prominences. Comparison of spatial expression patterns predicted a network of Wnt7a-associated GT-enriched epithelial genes, including Gjb2, Dsc3, Krt5, and Sostdc1. Known from other contexts, these genes are associated with normal epidermal differentiation, with disruptions in Dsc3 and Gjb2 showing palmo-plantar keratoderma in the limb. We propose that this gene network contributes to normal foreskin, scrotum, and labial development. As several of these genes are known to be regulated by, or contain cis elements responsive to retinoic acid, estrogen, or androgen, this implicates this pathway in the later androgen-dependent development of the GT.


Asunto(s)
Expresión Génica , Redes Reguladoras de Genes , Sistema Urogenital/embriología , Andrógenos/genética , Animales , Diferenciación Celular/genética , Embrión de Mamíferos , Epidermis , Extremidades , Genitales Masculinos/embriología , Masculino , Ratones , Organogénesis/genética , Uretra/embriología
18.
Development ; 137(13): 2177-86, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20530545

RESUMEN

In Drosophila melanogaster, dorsal closure is a model of tissue morphogenesis leading to the dorsal migration and sealing of the embryonic ectoderm. The activation of the JNK signal transduction pathway, specifically in the leading edge cells, is essential to this process. In a genome-wide microarray screen, we identified new JNK target genes during dorsal closure. One of them is the gene scarface (scaf), which belongs to the large family of trypsin-like serine proteases. Some proteins of this family, like Scaf, bear an inactive catalytic site, representing a subgroup of serine protease homologues (SPH) whose functions are poorly understood. Here, we show that scaf is a general transcriptional target of the JNK pathway coding for a secreted SPH. scaf loss-of-function induces defects in JNK-controlled morphogenetic events such as embryonic dorsal closure and adult male terminalia rotation. Live imaging of the latter process reveals that, like for dorsal closure, JNK directs the dorsal fusion of two epithelial layers in the pupal genital disc. Genetic data show that scaf loss-of-function mimics JNK over-activity. Moreover, scaf ectopic expression aggravates the effect of the JNK negative regulator puc on male genitalia rotation. We finally demonstrate that scaf acts as an antagonist by negatively regulating JNK activity. Overall, our results identify the SPH-encoding gene scaf as a new transcriptional target of JNK signalling and reveal the first secreted regulator of the JNK pathway acting in a negative-feedback loop during epithelial morphogenesis.


Asunto(s)
Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal , Animales , Embrión no Mamífero/metabolismo , Retroalimentación , Genitales Masculinos/embriología , Masculino , Morfogénesis
19.
J Pediatr Urol ; 6(4): 346-54, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19995686

RESUMEN

Hypospadias is one of the most common congenital disorders in males. Impaired fetal androgen action interferes with masculinization, including external genitalia formation, and can result in this anomaly; however, the molecular etiology remains unknown. Recent molecular approaches, including gene-targeting approaches in mice and single nucleotide polymorphisms analyses in humans, might provide an opportunity to identify the causative and risk factors of this anomaly. Several genes, such as sonic hedgehog, fibroblast growth factors, bone morphogenetic proteins, homeobox genes, and the Wnt family regulate external genitalia formation. Mastermind-like domain containing 1/chromosome X open reading frame 6 mutation and activating transcription factor 3 variants have been shown to be associated with the incidence of isolated hypospadias. In addition, this anomaly may be associated with a specific haplotype of the gene for estrogen receptor alpha, which mediates the estrogenic effects of environmental endocrine disruptors, and the effects of these disruptors on external genitalia formation might depend on individual genetic susceptibility. These molecular studies will refine our knowledge of the genetic mechanism involved in external genitalia formation, and lead to new strategies for the clinical management of hypospadias.


Asunto(s)
Genitales Masculinos/embriología , Hipospadias/genética , Animales , Hormonas Esteroides Gonadales/genética , Hormonas Esteroides Gonadales/fisiología , Humanos , Masculino , Diferenciación Sexual/genética
20.
Rev. chil. obstet. ginecol ; 75(2): 117-123, 2010. ilus, tab
Artículo en Español | LILACS | ID: lil-565387

RESUMEN

Objetivos: Valorar la precisión de la determinación ecográfíca del sexo fetal entre las 11 y las 14 semanas en una cohorte no seleccionada de gestantes. Métodos: Se realizó un estudio prospectivo transversal en una serie consecutiva de 636 gestaciones entre las 11 y 14 semanas, en gestantes que acudieron para el estudio ecográfco rutinario de primer trimestre. Se examinó la región genital del feto en un plano mediosagi-tal, visualizando el signo sagital y la dirección en la que apunta el tubérculo genital (craneal en los varones, caudal en las mujeres) ambos indicadores del sexo fetal. La confrmación clínica del sexo fetal se obtuvo después del parto por confrmación telefónica, o por el cariotipo en casos con amniocentesis por indicaciones convencionales. Resultados: Se consiguió asignar el sexo fetal correctamente en el 86,3 por ciento de los fetos. La precisión de la asignación ecográfca del sexo fetal fue mayor en varones que en mujeres (el 90,6 por ciento de los varones fueron asignados correctamente vs el 83,1 por ciento de las mujeres, p<0,05), y se incrementó con la edad gestacional desde un 72 por ciento entre las 11 y 11+6 semanas, un 92 por ciento entre las 12 y las 12+6 semanas, hasta un 95 por ciento entre las 13 y las 13+6 semanas. Conclusión: La determinación ecográfíca del sexo fetal tiene una elevada tasa de precisión a partir de las 13 semanas, lo que sugiere que las pruebas invasivas para la exclusión de enfermedades ligadas al X podrían obviarse cuando se identifca un feto varón a esta edad gestacional. Sin embargo, en fetos identifcados como mujeres esta decisión deberá posponerse hasta una edad gestacional más avanzada.


Objective: To assess the accuracy of fetal gender determination by ultrasound at 11-14 weeks of gestation in a large cohort of unselected population. Methods: A prospective cross-sectional study was performed in 636 consecutive pregnancies at 11-14 weeks of gestation, attending for frst trimester ultrasound screening. The genital region was examined in the mid-sagittal plane to detect the sagittal sign and the direction in which the genital tubercle pointed (cranial for males, caudal for females) both as markers of fetal gender. The clinical confrmation of fetal gender was obtained from telephonic interviews after delivery or from karyotype in cases scheduled to amniocentesis for conventional indications. Results: Fetal gender was correctly determined by ultrasound in 86.3 percent of the fetuses. The accuracy of sex assignment was higher in male fetuses than in female fetuses (90.6 percent of the males fetuses vs 83.1 percent of the females were correctly assigned, p<0.05) and increased with gestational age from 72 percent at 11-11+6 week's to 92 percent at 12-12+6 week's, and 95 percent at 13-13+6 week's gestation (p<0.01). Conclusion: Prenatal gender assignment by ultrasound has a high accuracy rate at 13 to 13+6 weeks. These results suggest that invasive testing can probably be performed in fetuses identifed as males at this gestational age. However in fetuses identifed as females, the decision regarding invasive testing should be postponed until a higher gestational age is achieved.


Asunto(s)
Humanos , Análisis para Determinación del Sexo , Ultrasonografía Prenatal/métodos , Estudios Transversales , Genitales Femeninos/embriología , Genitales Femeninos , Genitales Masculinos/embriología , Genitales Masculinos , Primer Trimestre del Embarazo , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA