Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
FEBS Open Bio ; 14(1): 37-50, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37953493

RESUMEN

Male and female reproductive tracts develop from anterior intermediate mesoderm with similar differentiation processes. The anterior intermediate mesoderm develops into the mesonephros, and the Wolffian duct initiates by epithelialization in the mesonephros. The Müllerian duct invaginates from the coelomic epithelium of the cranial mesonephros for ductal formation and is then regionalized into proximal to caudal female reproductive tracts. In this study, we focused on the epithelialization of the Wolffian duct, initiation of the Müllerian duct, and the regionalization step of the Müllerian ducts as a continuous process. By using intermediate mesodermal cells from mouse pluripotent stem cells, we identified that inhibition of SMAD2/3 signaling might be involved in the differentiation into mesenchymal cells, after which mesonephric cells might be then epithelialized during differentiation of the Wolffian duct. Aggregation of coelomic epithelial cells might be related to initiation of the Müllerian duct. Transcriptomic analysis predicted that consensus sequences of SMAD3/4 were enriched among highly expressed genes in the proximal Müllerian duct. SMAD2/3 signaling to regulate differentiation of the Wolffian duct was continuously activated in the proximal Müllerian duct and was involved in proximal and oviductal regionalization. Therefore, SMAD2/3 signaling may be finely tuned to regulate differentiation from initiation to regionalization steps.


Asunto(s)
Conductos Paramesonéfricos , Conductos Mesonéfricos , Ratones , Animales , Masculino , Femenino , Conductos Mesonéfricos/fisiología , Conductos Paramesonéfricos/fisiología , Diferenciación Celular , Células Epiteliales , Transducción de Señal
2.
Curr Top Dev Biol ; 148: 51-77, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35461568

RESUMEN

In eutherian and marsupial mammals, the site of embryo implantation and gestation is the uterus. Uterine morphologies vary between mammalian species. For example, laboratory mice have a bipartite uterus with two uterine horns and a single cervix, whereas humans have a simplex uterus with a single chamber and single cervix. The precursor tissue of the uterus, oviducts, and upper vagina is the Müllerian duct epithelium and its adjacent mesenchyme. Morphological variation between species is established during embryogenesis by species-specific differences in Müllerian duct fusion at the midline, growth, and differentiation. In humans, alterations in Müllerian duct development can lead to variations in uterine morphology that correlate with increased risks of miscarriage and infertility. Here we review the developmental genetic factors that regulate Müllerian duct development, uterine morphogenesis, and human uterine variation.


Asunto(s)
Conductos Paramesonéfricos , Útero , Animales , Epitelio , Femenino , Mamíferos/genética , Ratones , Conductos Paramesonéfricos/fisiología , Organogénesis/genética , Útero/fisiología
3.
Annu Rev Anim Biosci ; 7: 149-172, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30303691

RESUMEN

In vertebrates, sex organs are generally specialized to perform a male or female reproductive role. Acquisition of the Müllerian duct, which gives rise to the oviduct, together with emergence of the Amh/Amhr2 system favored evolution of viviparity in jawed vertebrates. Species with high sex-specific reproductive adaptations have less potential to sex reverse, making intersex a nonfunctional condition. Teleosts, the only vertebrate group in which hermaphroditism evolved as a natural reproductive strategy, lost the Müllerian duct during evolution. They developed for gamete release complete independence from the urinary system, creating optimal anatomic and developmental preconditions for physiological sex change. The common and probably ancestral role of Amh is related to survival and proliferation of germ cells in early and adult gonads of both sexes rather than induction of Müllerian duct regression. The relationship between germ cell maintenance and sex differentiation is most evident in species in which Amh became the master male sex-determining gene.


Asunto(s)
Trastornos del Desarrollo Sexual/genética , Genoma/genética , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Diferenciación Sexual/genética , Transducción de Señal/genética , Vertebrados/genética , Animales , Evolución Biológica , Femenino , Gónadas/fisiología , Masculino , Conductos Paramesonéfricos/fisiología , Filogenia , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Vertebrados/fisiología
4.
Proc Natl Acad Sci U S A ; 115(33): 8382-8387, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30061417

RESUMEN

In mammals, the developing reproductive tract primordium of male and female fetuses consists of the Wolffian duct and the Müllerian duct (MD), two epithelial tube pairs surrounded by mesenchyme. During male development, mesenchyme-epithelia interactions mediate MD regression to prevent its development into a uterus, oviduct, and upper vagina. It is well established that transforming growth factor-ß family member anti-Müllerian hormone (AMH) secreted from the fetal testis and its type 1 and 2 receptors expressed in MD mesenchyme regulate MD regression. However, little is known about the molecular network regulating downstream actions of AMH signaling. To identify potential AMH-induced genes and regulatory networks controlling MD regression in a global nonbiased manner, we examined transcriptome differences in MD mesenchyme between males (AMH signaling on) and females (AMH signaling off) by RNA-seq analysis of purified fetal MD mesenchymal cells. This analysis found 82 genes up-regulated in males during MD regression and identified Osterix (Osx)/Sp7, a key transcriptional regulator of osteoblast differentiation and bone formation, as a downstream effector of AMH signaling during MD regression. Osx/OSX was expressed in a male-specific pattern in MD mesenchyme during MD regression. OSX expression was lost in mutant males without AMH signaling. In addition, transgenic mice ectopically expressing human AMH in females induced a male pattern of Osx expression. Together, these results indicate that AMH signaling is necessary and sufficient for Osx expression in the MD mesenchyme. In addition, MD regression was delayed in Osx-null males, identifying Osx as a factor that regulates MD regression.


Asunto(s)
Hormona Antimülleriana/fisiología , Conductos Paramesonéfricos/fisiología , Transducción de Señal/fisiología , Factor de Transcripción Sp7/fisiología , Animales , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , beta Catenina/fisiología
5.
Fertil Steril ; 103(2): 487-93, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25492685

RESUMEN

OBJECTIVE: To examine whether pathogenic copy number changes (CNCs) can be identified in deoxyribonucleic acid from females with different classes of müllerian anomalies. DESIGN: We conducted array-based copy number variant (CNV) analysis using an oligonucleotide array from deoxyribonucleic acid in 12 adolescent females with various müllerian anomalies. SETTING: University-affiliated tertiary care institution. PATIENT(S): Twenty adolescent females with clinically confirmed müllerian anomalies. INTERVENTION(S): Array-based CNV analysis. MAIN OUTCOME MEASURE(S): Copy number changes and/or regions with absence of heterozygosity. RESULT(S): A total of 192 CNVs identified in these samples were previously annotated as polymorphic. Three CNCs that were identified in regions with minimal to no overlap with annotated polymorphisms failed significance criteria with detailed inspection. One subject harbored a 5.1-Mb region of absence of heterozygosity at Xq23 that is of unknown significance. CONCLUSION(S): We did not identify pathogenic CNCs in this small pilot cohort of patients with various müllerian anomalies, but larger studies will be needed to further investigate whether CNCs are associated with all classes of müllerian anomalies.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Estudio de Asociación del Genoma Completo/métodos , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/fisiología , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Niño , Estudios de Cohortes , Femenino , Humanos , Proyectos Piloto , Adulto Joven
6.
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
7.
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
8.
Diabetes ; 59(9): 2297-305, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20530741

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF-A or VEGF) is a major pathogenic factor and therapeutic target for diabetic retinopathy (DR). Since VEGF has been proposed as a survival factor for retinal neurons, defining the cellular origin of pathogenic VEGF is necessary for the effectiveness and safety of long-term anti-VEGF therapies for DR. To determine the significance of Müller cell-derived VEGF in DR, we disrupted VEGF in Müller cells with an inducible Cre/lox system and examined diabetes-induced retinal inflammation and vascular leakage in these conditional VEGF knockout (KO) mice. RESEARCH DESIGN AND METHODS: Leukostasis was determined by counting the number of fluorescently labeled leukocytes inside retinal vasculature. Expression of biomarkers for retinal inflammation was assessed by immunoblotting of TNF-alpha, ICAM-1, and NF-kappaB. Vascular leakage was measured by immunoblotting of retinal albumin and fluorescent microscopic analysis of extravascular albumin. Diabetes-induced vascular alterations were examined by immunoblotting and immunohistochemistry for tight junctions, and by trypsin digestion assays for acellular capillaries. Retinal integrity was analyzed with morphologic and morphometric analyses. RESULTS: Diabetic conditional VEGF KO mice exhibited significantly reduced leukostasis, expression of inflammatory biomarkers, depletion of tight junction proteins, numbers of acellular capillaries, and vascular leakage compared to diabetic control mice. CONCLUSIONS: Müller cell-derived VEGF plays an essential and causative role in retinal inflammation, vascular lesions, and vascular leakage in DR. Therefore, Müller cells are a primary cellular target for proinflammatory signals that mediates retinal inflammation and vascular leakage in DR.


Asunto(s)
Retinopatía Diabética/fisiopatología , Conductos Paramesonéfricos/fisiología , Retina/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Peso Corporal , Capilares/patología , Capilares/fisiopatología , Cartilla de ADN , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación , Leucostasis/patología , Leucostasis/fisiopatología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Vasos Retinianos/patología , Vasos Retinianos/fisiopatología , Uniones Estrechas/fisiología , Factor A de Crecimiento Endotelial Vascular/deficiencia , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/uso terapéutico
9.
Ann Endocrinol (Paris) ; 71(3): 191-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20362961

RESUMEN

In this review, the role of anti-Müllerian hormone (AMH) as a regulator and marker of ovarian function is described. Studies in mice showed that AMH is one of the intra-ovarian growth factors regulating primordial follicle recruitment and follicle-stimulating hormone (FSH) sensitivity of growing follicles in an inhibitory manner. Association studies of common variants of the AMH and AMHRII gene suggest that AMH may have a similar role in the human ovary. When it was discovered that serum levels AMH are correlated with the number of growing follicles, AMH gained further clinical interest as a marker for the quantitative aspect of ovarian reserve and as a diagnostic marker for polycystic ovary syndrome (PCOS).


Asunto(s)
Hormona Antimülleriana/fisiología , Ovario/fisiología , Animales , Hormona Antimülleriana/deficiencia , Hormona Antimülleriana/genética , Femenino , Genotipo , Homeostasis , Humanos , Ratones , Ratones Noqueados , Conductos Paramesonéfricos/fisiología , Folículo Ovárico/fisiología , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética
10.
Reproduction ; 139(4): 749-57, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20089665

RESUMEN

The somatic (Sertoli cell (SC), Leydig cell (LC), and peritubular myoid (PTM) cell) cells play key roles in development of the fetal testis. We established monolayer cultures from second trimester human testes and investigated the pattern of expression of cell-lineage characteristic mRNAs. Expression of some SC-associated genes (SRY, SOX9, WT1, GATA4, and SF1) was detectable up to and including passage 3 (P3), while others (anti-Müllerian hormone; desert hedgehog) present prior to dissociation were not expressed in the cultured cells. Transcripts encoding the androgen receptor were expressed but addition of dihydrotestosterone (DHT) had no impact on expression of mRNAs expressed in SC or LC. Total concentrations of mRNAs encoding smooth muscle actin (ACTA2) and desmin increased from P1 to P3; an increasing proportion of the cells in the cultures were immunopositive for ACTA2 consistent with proliferation/differentiation of PTM cells. In conclusion, somatic cell monolayer cultures were established from human fetal testes; these cultures could form the basis for future studies based on isolation of purified populations of somatic cells and manipulation of gene expression that is difficult to achieve with organ culture systems. Our results suggest that fetal SC do not maintain a fully differentiated phenotype in vitro, yet PTM (ACTA2 positive) cells readily adapt to monolayer culture conditions in the presence of DHT. This culture system provides an opportunity to study the impact of regulatory factors on gene expression in PTM cells, a population thought to play a key role in mediating androgen action within the developing testis.


Asunto(s)
Andrógenos/farmacología , Proliferación Celular/efectos de los fármacos , Feto/citología , Células de Sertoli/citología , Células de Sertoli/fisiología , Testículo/embriología , Biomarcadores/análisis , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Femenino , Feto/fisiología , Humanos , Masculino , Conductos Paramesonéfricos/citología , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/fisiología , Especificidad de Órganos , Embarazo , Segundo Trimestre del Embarazo , Testículo/citología , Testículo/fisiología , Estudios de Validación como Asunto
11.
Fertil Steril ; 94(1): 120-5, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19324347

RESUMEN

OBJECTIVE: To investigate whether Y chromosomal genetic material has a role in the development of Müllerian aplasia in Finland. We have studied the TSPY1 gene and 38 additional male-specific fragments covering areas of both the long and short arms of the Y chromosome in Finnish patients with Müllerian aplasia. DESIGN: A retrospective study. SETTING: University hospital and genetic laboratory. PATIENT(S): A sample set of 110 Finnish patients with well-diagnosed Müllerian aplasia and 20 healthy relatives (13 mothers, 4 fathers, and 3 sisters from different families) were included in the study. One hundred healthy female controls with a background of at least one normal pregnancy with delivery were used as controls. INTERVENTION(S): Blood samples for DNA extraction. MAIN OUTCOME MEASURE(S): Detection of Y chromosomal fragments by polymerase chain reaction in female patients with Müllerian aplasia. RESULT(S): None of the female patients showed presence of the earlier reported TSPY1 gene or 38 additional Y chromosomal markers. CONCLUSION(S): Our results indicate that the studied Y-specific fragments, namely TSPY1 and 38 Y chromosomal markers, are not responsible for the syndrome in these Finnish patients with Müllerian aplasia.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cromosomas Humanos Y/fisiología , Conductos Paramesonéfricos/anomalías , Femenino , Finlandia , Marcadores Genéticos/genética , Humanos , Masculino , Conductos Paramesonéfricos/fisiología , Estudios Retrospectivos , Síndrome
12.
J Pediatr Surg ; 44(12): 2335-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20006021

RESUMEN

AIM: Mullerian-inhibiting substance (MIS) may have a role in postnatal germ cell development, although this remains unproven. Elucidating the regulatory factors is crucial in finding new treatments for preventing infertility in cryptorchidism. We studied germ cell development in neonatal mice with MIS gene or receptor mutation to determine if germ cell development was affected. METHODS: Neonatal (5 MIS mutants, x1 MIS receptor mutant and 5 wild-type) and 10-day-old mice (x 7 MIS mutants, x1 MIS receptor mutant, 5 wild-type) were killed and prepared for hematoxylin-eosin and Masson trichrome histology of the testis. Testis diameter and tubule diameter were measured by Image-J, and germ cells were counted in 50 tubules/testis. RESULTS: Total testis and tubular diameters were greater in wild-type vs MIS mutants at days 0 and 10 (P < .01). Gonocytes were decreased in MIS mutants vs wild-type on day 0 (P = .019), and on day 10, the number of type A spermatogonia was slightly decreased (P = .05) and type B spermatogonia were significantly decreased (P < .01). Similar results were seen in the MIS receptor knockout. CONCLUSION: These results suggest that MIS has a previously unrecognized role in perinatal germ cell development that needs further investigation. Mullerian-inhibiting substance may be a possible future treatment for stimulating germ cell development in cryptorchidism.


Asunto(s)
Hormona Antimülleriana/deficiencia , Criptorquidismo/complicaciones , Células Germinativas/crecimiento & desarrollo , Infertilidad Masculina/etiología , Animales , Animales Recién Nacidos , Hormona Antimülleriana/genética , Hormona Antimülleriana/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Criptorquidismo/genética , Criptorquidismo/fisiopatología , Modelos Animales de Enfermedad , Células Madre Embrionarias/fisiología , Infertilidad Masculina/genética , Infertilidad Masculina/fisiopatología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Conductos Paramesonéfricos/fisiología , Orquidopexia/métodos , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Espermatogonias/crecimiento & desarrollo , Espermatogonias/fisiología , Testículo/embriología , Testículo/crecimiento & desarrollo
13.
Trends Endocrinol Metab ; 20(7): 357-63, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19709900

RESUMEN

Growing concerns over endocrine disrupting chemicals (EDCs) and their effects on human fetal development and adult health have promoted research into the underlying molecular mechanisms of endocrine disruption. Gene targeting technology has allowed insight into the genetic pathways governing reproductive tract development and how exposure to EDCs during a critical developmental window can alter reproductive tract development, potentially forming the basis for adult diseases. This review primarily uses diethylstilbestrol (DES) as a model agent for EDCs and discusses the recent progress elucidating how DES and other EDCs affect murine female reproductive tract development and cancer at the molecular level.


Asunto(s)
Dietilestilbestrol/farmacología , Disruptores Endocrinos/farmacología , Genitales Femeninos/efectos de los fármacos , Conductos Paramesonéfricos/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Genitales Femeninos/crecimiento & desarrollo , Genitales Femeninos/fisiología , Humanos , Ratones , Conductos Paramesonéfricos/fisiología , Embarazo
14.
Genesis ; 46(4): 205-13, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18395837

RESUMEN

dachshund/Dach gene family members encode transcriptional cofactors with highly conserved protein interaction domains and are expressed in the developing eyes, brains, and limbs in insects and vertebrates. These observations suggest that the developmental roles of dachshund/Dach in these tissues have been conserved since the divergence of arthropods and chordates. However, while Drosophila dachshund mutants have abnormalities in eye, brain, limbs, mouse Dach1 or Dach2 knockout mutants do not exhibit gross anatomical malformations in these tissues. In addition, Dach1/2 double homozygotes have intact eyes and limbs. Here we show that in Dach1/Dach2 double mutants, female reproductive tract (FRT) development is severely disrupted. This defect is associated with the Müllerian duct (MD) and not the Wolffian duct (WD), which normally differentiate into either the FRT or male reproductive tract (MRT), respectively. Dach1 and Dach2 are expressed in the MD, and in Dach1/2 double mutants, MD expression of Lim1 and Wnt7a is abnormal and MD development is disrupted. In contrast, WD and MRT development are not grossly affected. We propose that Dach1 and Dach2 proteins may redundantly control FRT formation by regulating the expression of target genes required for development of the MD. This vertebrate Dach1/2 function may have been conserved during arthropod evolution, as Drosophila dachshund mutants also exhibit an FRT phenotype.


Asunto(s)
Proteínas del Ojo/fisiología , Conductos Paramesonéfricos/embriología , Proteínas Nucleares/fisiología , Animales , Proteínas de Unión al ADN , Drosophila/embriología , Drosophila/genética , Drosophila/fisiología , Proteínas del Ojo/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Conductos Paramesonéfricos/fisiología , Proteínas Nucleares/genética , Factores de Transcripción , Conductos Mesonéfricos/embriología , Conductos Mesonéfricos/fisiología
15.
Rev Med Brux ; 28(3): 165-71, 2007.
Artículo en Francés | MEDLINE | ID: mdl-17708472

RESUMEN

Anti-mullerian hormone, also called AMH, belongs to the large family of transforming growth factor P. Its role in the sexual differentiation of male fetus is now well known. Recently, AMH has been demonstrated to play an important role in the ovarian function. In fact, AMH seems to regulate the kinetics of follicular development, inhibiting the follicular recruitment and the follicular growth. Thus, this intra-gonadic cybernin could be a decisive determinant of the rapidity of follicular pool exhaustion. Today, some experimental data from the literature suggest that AMH could be a reliable marker of ovarian reserve. This review summarizes the present knowledge about AMH and its role in physiology but also in ovarian pathology.


Asunto(s)
Glicoproteínas/fisiología , Ovario/fisiología , Hormonas Testiculares/fisiología , Hormona Antimülleriana , Biomarcadores/análisis , Femenino , Glicoproteínas/análisis , Hormonas Esteroides Gonadales/fisiología , Humanos , Conductos Paramesonéfricos/fisiología , Neuropéptidos/fisiología , Enfermedades del Ovario/diagnóstico , Folículo Ovárico/fisiología , Hormonas Testiculares/análisis , Factor de Crecimiento Transformador beta/fisiología
16.
Anat Histol Embryol ; 36(4): 272-83, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17617105

RESUMEN

In this study the development of the bovine Fallopian tube was investigated using light microscopic methods. Formation and differentiation of the Müllerian duct were studied in mesonephroi of 16 embryos and fetuses with a crown-rump lengths (CRL) of 0.9-8.4 cm. The funnel field, the rostral beginning of the Müllerian duct was first observed at a CRL of 0.9 cm. It appears as a thickening of the mesothelium on the craniolateral side of the mesonephros. During later development the Müllerian duct emerges by caudal outgrowth from the funnel field. Formation of a common basal lamina surrounding the caudal tips of Müllerian and Wolffian ducts could be observed at all stages up to CRL of 2.7 cm. The mesothelium and the epithelium of the Wolffian duct adjacent to the Müllerian duct showed a modification of epithelium height in all examined stages. Probably the Wolffian duct influences the growth of Müllerian duct by epithelio-mesenchymal interactions. Fetuses from a CRL of 12.0 to 94.0 cm were used for investigation of the prenatal differentiation of the oviductal mucosa. Folding of the oviductal mucosa started at a CRL of 29.0 cm and continued until birth. Individual primary, secondary and tertiary folds are formed in special proliferation zones and epithelium-folding buds. The cellular differentiation of the oviductal epithelium involves the formation of ciliated and secretory cells during different times of prenatal development. Ciliogenesis was first detected at a CRL of 33.0 cm. Active secretory cells could be observed in the oviductal epithelium from a CRL of 64.0 cm onwards.


Asunto(s)
Bovinos/anatomía & histología , Bovinos/embriología , Trompas Uterinas/embriología , Animales , Trompas Uterinas/anatomía & histología , Trompas Uterinas/fisiología , Femenino , Masculino , Conductos Paramesonéfricos/anatomía & histología , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/fisiología , Conductos Mesonéfricos/anatomía & histología , Conductos Mesonéfricos/embriología , Conductos Mesonéfricos/fisiología
17.
Sex Dev ; 1(2): 100-10, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18391520

RESUMEN

The development of a vagina as a separate outlet of the birth canal evolves at the transition of egg laying species to eutherian mammals. The derivation of the vagina from the Wolffian and Müllerian ducts and the contribution of the urogenital sinus are still open questions. Here experiments with the complete androgen receptor defect in the testicular feminisation (Tfm) mouse are reported which show that the vagina is formed by caudal migration of Wolffian and Müllerian ducts. The cranial ends of the Wolffian ducts successively regress while the Müllerian ducts fuse to form the vagina. Immunohistochemistry of the androgen receptor reveals that the caudal ends of the Wolffian ducts remain in the indifferent stage and therefore have been mistaken as sinuvaginal bulbs. The Wolffian ducts do not contribute to the vagina itself but have a helper function during downward movement of the vaginal bud in the female. In the male the caudal ends serve as androgen operated switch for the negative control of vaginal development. The results indicate that the rudimentary vagina in the complete androgen insensitivity syndrome (CAIS) corresponds to non obliterated caudal ends of the Müllerian ducts. Selective atresia of the vagina in the MRKH (Mayer-Rokitansky-Kuster-Hauser) syndrome may be explained by the failure of Wolffian and Müllerian ducts to descend caudally.


Asunto(s)
Andrógenos/fisiología , Vagina/anatomía & histología , Conductos Mesonéfricos/fisiología , Andrógenos/metabolismo , Animales , Femenino , Heterocigoto , Inmunohistoquímica , Masculino , Ratones , Ratones Mutantes , Modelos Biológicos , Conductos Paramesonéfricos/anatomía & histología , Conductos Paramesonéfricos/fisiología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Vagina/embriología , Vagina/metabolismo , Conductos Mesonéfricos/anatomía & histología
18.
Sex Dev ; 1(5): 271-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18391537

RESUMEN

Several recent publications have contributed to our understanding of the processes involved in development of the Müllerian ducts in both sexes and regression of these structures in male embryos. Additionally, new insights in the regulation of the anti-Müllerian hormone (AMH) signaling pathway, the pathway, which mediates the male specific degeneration of Müllerian ducts, have been gained. It has become clear that the Müllerian duct is formed by invagination of the coelomic epithelium and elongates primarily by proliferation. Later on cells of the coelomic epithelium perform epithelial to mesenchymal transition and move around the epithelium of the Müllerian duct to induce degeneration of this structure in male embryos. Besides AMH and its specific type II receptor AMHR2 two different type I receptors as well as different SMAD family members have been shown to be involved in the AMH signaling cascade. Other factors including WT1, WNT7a, beta-catenin and MMP2 act upstream and downstream of AMH signaling. Here we try to draw an overall picture of Müllerian duct formation and regression by integrating the recent literature in the field.


Asunto(s)
Conductos Paramesonéfricos/crecimiento & desarrollo , Conductos Paramesonéfricos/fisiología , Animales , Hormona Antimülleriana/fisiología , Femenino , Masculino , Conductos Paramesonéfricos/anatomía & histología , Transducción de Señal/fisiología
19.
Development ; 133(12): 2359-69, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16687449

RESUMEN

Examination of Müllerian inhibiting substance (MIS) signaling in the rat in vivo and in vitro revealed novel developmental stage- and tissue-specific events that contributed to a window of MIS responsiveness in Müllerian duct regression. The MIS type II receptor (MISRII)-expressing cells are initially present in the coelomic epithelium of both male and female urogenital ridges, and then migrate into the mesenchyme surrounding the male Müllerian duct under the influence of MIS. Expression of the genes encoding MIS type I receptors, Alk2 and Alk3, is also spatiotemporally controlled; Alk2 expression appears earlier and increases predominantly in the coelomic epithelium, whereas Alk3 expression appears later and is restricted to the mesenchyme, suggesting sequential roles in Müllerian duct regression. MIS induces expression of Alk2, Alk3 and Smad8, but downregulates Smad5 in the urogenital ridge. Alk2-specific small interfering RNA (siRNA) blocks both the transition of MISRII expression from the coelomic epithelium to the mesenchyme and Müllerian duct regression in organ culture. Müllerian duct regression can also be inhibited or accelerated by siRNA targeting Smad8 and Smad5, respectively. Thus, the early action of MIS is to initiate an epithelial-to-mesenchymal transition of MISRII-expressing cells and to specify the components of the receptor/SMAD signaling pathway by differentially regulating their expression.


Asunto(s)
Células Epiteliales/fisiología , Glicoproteínas/metabolismo , Mesodermo/fisiología , Conductos Paramesonéfricos/fisiología , Receptores de Péptidos/metabolismo , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Hormonas Testiculares/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Hormona Antimülleriana , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Movimiento Celular/fisiología , Estructuras Embrionarias/anatomía & histología , Estructuras Embrionarias/fisiología , Células Epiteliales/citología , Femenino , Glicoproteínas/genética , Humanos , Hibridación in Situ , Masculino , Mesodermo/citología , Ratones , Conductos Paramesonéfricos/anatomía & histología , Embarazo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Smad/genética , Hormonas Testiculares/genética
20.
Fertil Steril ; 84(4): 938-44, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16213847

RESUMEN

OBJECTIVE: To compare serum müllerian-inhibiting substance (MIS) concentrations in adolescent girls with polycystic ovary syndrome (PCOS) or normal menstrual cycles. DESIGN: Prospective study. SETTING: University department of obstetrics and gynecology. PATIENT(S): Thirty-one girls (12-18 years old) with PCOS and 17 girls (12-19 years old) with normal menstrual cycles. INTERVENTION(S): Serum was collected from girls with PCOS or normal cycles during the early follicular phase of the menstrual cycle, stored frozen until assayed. MAIN OUTCOME MEASURE(S): Serum levels of MIS, E2, free-T, androstenedione, LH, and FSH. RESULT(S): Serum MIS levels in girls with PCOS were significantly higher compared with normal girls (4.1 +/- 2.2 [SD] and 2.4 +/- 1.0 ng/mL, respectively). The subjects were stratified for body mass index (BMI) (< and >25 kg/m2). Serum MIS levels in PCOS girls (4.2 +/- 3.0 ng/mL [BMI <25 kg/m2] and 4.0 +/- 1.6 [BMI >25 kg/m2]) were significantly higher compared with normal girls (2.2 +/- 0.8 and 2.6 +/- 0.7 ng/mL, respectively). CONCLUSION(S): Adolescent girls with PCOS have significantly higher serum MIS levels compared with normally cycling girls. Serum MIS levels in PCOS were not influenced by BMI. Increased MIS production may represent an early manifestation of the disease.


Asunto(s)
Glicoproteínas/sangre , Ciclo Menstrual/sangre , Conductos Paramesonéfricos/fisiología , Síndrome del Ovario Poliquístico/sangre , Hormonas Testiculares/sangre , Adolescente , Adulto , Hormona Antimülleriana , Biomarcadores/sangre , Niño , Femenino , Humanos , Conductos Paramesonéfricos/patología , Síndrome del Ovario Poliquístico/fisiopatología , Estudios Prospectivos , Texas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...