Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biol Reprod ; 99(4): 735-748, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29688260

RESUMEN

Despite the major negative impact uterine fibroids (UFs) have on female reproductive health, little is known about early events that initiate development of these tumors. Somatic fibroid-causing mutations in mediator complex subunit 12 (MED12), the most frequent genetic alterations in UFs (up to 85% of tumors), are implicated in transforming normal myometrial stem cells (MSCs) into tumor-forming cells, though the underlying mechanism(s) leading to these mutations remains unknown. It is well accepted that defective DNA repair increases the risk of acquiring tumor-driving mutations, though defects in DNA repair have not been explored in UF tumorigenesis. In the Eker rat UF model, a germline mutation in the Tsc2 tumor suppressor gene predisposes to UFs, which arise due to "second hits" in the normal allele of this gene. Risk for developing these tumors is significantly increased by early-life exposure to endocrine-disrupting chemicals (EDCs), suggesting increased UF penetrance is modulated by early drivers for these tumors. We analyzed DNA repair capacity using analyses of related gene and protein expression and DNA repair function in MSCs from adult rats exposed during uterine development to the model EDC diethylstilbestrol. Adult MSCs isolated from developmentally exposed rats demonstrated decreased DNA end-joining ability, higher levels of DNA damage, and impaired ability to repair DNA double-strand breaks relative to MSCs from age-matched, vehicle-exposed rats. These data suggest that early-life developmental EDC exposure alters these MSCs' ability to repair and reverse DNA damage, providing a driver for acquisition of mutations that may promote the development of these tumors in adult life.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Leiomioma/etiología , Miometrio/efectos de los fármacos , Miometrio/metabolismo , Neoplasias Uterinas/etiología , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/metabolismo , Animales , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Daño del ADN , Reparación del ADN/genética , Dietilestilbestrol/toxicidad , Modelos Animales de Enfermedad , Femenino , Mutación de Línea Germinal , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Complejo Mediador/genética , Miometrio/crecimiento & desarrollo , Ratas , Ratas Mutantes , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo
2.
Reprod Fertil Dev ; 27(7): 985-99, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24630125

RESUMEN

Female mice lacking the follistatin gene but expressing a human follistatin-315 transgene (tghFST315) have reproductive abnormalities (reduced follicles, no corpora lutea and ovarian-uterine inflammation). We hypothesised that the absence of follistatin-288 causes the abnormal reproductive tract via both developmental abnormalities and abnormal ovarian activity. We characterised the morphology of oviducts and uteri in wild type (WT), tghFST315 and follistatin-knockout mice expressing human follistatin-288 (tghFST288). The oviducts and uteri were examined in postnatal Day-0 and adult mice (WT and tghFST315 only) using histology and immunohistochemistry. Adult WT and tghFST315 mice were ovariectomised and treated with vehicle, oestradiol-17ß (100ng injection, dissection 24h later) or progesterone (1mg×three daily injections, dissection 24h later). No differences were observed in the oviducts or uteri at birth, but abnormalities developed by adulthood. Oviducts of tghFST315 mice failed to coil, the myometrium was disorganised, endometrial gland number was reduced and oviducts and uteri contained abundant leukocytes. After ovariectomy, tghFST315 mice had altered uterine cell proliferation, and inflammation was maintained and exacerbated by oestrogen. These studies show that follistatin is crucial to postnatal oviductal-uterine development and function. Further studies differentiating the role of ovarian versus oviductal-uterine follistatin in reproductive tract function at different developmental stages are warranted.


Asunto(s)
Folistatina/genética , Oviductos/crecimiento & desarrollo , Útero/crecimiento & desarrollo , Animales , Proliferación Celular/genética , Endometrio/crecimiento & desarrollo , Endometrio/metabolismo , Estrógenos/farmacología , Femenino , Folistatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Miometrio/crecimiento & desarrollo , Miometrio/metabolismo , Ovariectomía , Oviductos/diagnóstico por imagen , Oviductos/metabolismo , Útero/efectos de los fármacos , Útero/metabolismo
3.
PLoS Genet ; 7(8): e1002210, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21852953

RESUMEN

T-box proteins are conserved transcription factors that play crucial roles in development of all metazoans; and, in humans, mutations affecting T-box genes are associated with a variety of congenital diseases and cancers. Despite the importance of this transcription factor family, very little is known regarding how T-box factors regulate gene expression. The Caenorhabditis elegans genome contains 21 T-box genes, and their characterized functions include cell fate specification in a variety of tissues. The C. elegans Tbx1 sub-family member MLS-1 functions during larval development to specify the fate of non-striated uterine muscles; and, in mls-1 mutants, uterine muscles are transformed to a vulval muscle fate. Here we demonstrate that MLS-1 function depends on binding to the Groucho-family co-repressor UNC-37. MLS-1 interacts with UNC-37 via a conserved eh1 motif, and the MLS-1 eh1 motif is necessary for MLS-1 to specify uterine muscle fate. Moreover, unc-37 loss-of-function produces uterine muscle to vulval muscle fate transformation similar to those observed in mls-1 mutants. Based on these results, we conclude that MLS-1 specifies uterine muscle fate by repressing target gene expression, and this function depends on interaction with UNC-37. Moreover, we suggest that MLS-1 shares a common mechanism for transcriptional repression with related T-box factors in other animal phyla.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Miometrio/crecimiento & desarrollo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Animales , Tipificación del Cuerpo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Organismos Hermafroditas , Mutación , Miometrio/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Dominio T Box/química , Proteínas de Dominio T Box/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos
4.
J Agric Food Chem ; 69(5): 1693-1704, 2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33528250

RESUMEN

Genistein is naturally occurring in plants and binds to estrogen receptors. Humans are mainly exposed through diet, but the use of supplements is increasing as genistein is claimed to promote health and alleviate menopausal symptoms. We analyzed diverse uterine features in adult mice chronically fed genistein for different times. The luminal epithelium height was increased in females treated with 500 and 1000 ppm at PND 95, and the width of the outer myometrium was increased in females treated with 1000 ppm at PND 65 compared to that in controls. An increase in proliferation was noted in the inner myometrium layer of animals exposed to 300 ppm genistein at PND 185 compared to that in controls. Luminal hyperplasia was greater in the 1000 ppm group at PND 65, 95, and 185, although not statistically different from control. These results indicate that genistein may exert estrogenic activity in the uterus, without persistent harm to the organ.


Asunto(s)
Genisteína/farmacología , Fitoestrógenos/farmacología , Útero/efectos de los fármacos , Útero/crecimiento & desarrollo , Animales , Proliferación Celular/efectos de los fármacos , Exposición Dietética , Femenino , Ratones , Miometrio/efectos de los fármacos , Miometrio/crecimiento & desarrollo
5.
Reproduction ; 139(6): 1067-75, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20368191

RESUMEN

We previously demonstrated that in the CD-1 mouse, which exhibits a high incidence of age-related adenomyosis, neonatal exposure to tamoxifen induced premature uterine adenomyosis and was associated with abnormal development particularly of the inner myometrium. In the present study, we examined the effect of neonatal tamoxifen administration upon uterine development in the C57/BL6J mouse strain that is not known to develop uterine adenomyosis. Female C57/BL6J pups (n=20) were treated with oral tamoxifen (1 mg/kg) from age 1 to 5 days. Uteri from control and treated mice were obtained on days 5, 10, 15 and 42 of age. We examined sections histologically using image analysis and immunohistochemistry for alpha-smooth muscle actin (ACTA2, alpha-SMA), desmin, vimentin, laminin, fibronectin and oestrogen receptor-alpha (ESR1). Following tamoxifen exposure, all uteri showed inner myometrium thinning, lack of continuity, disorganisation and bundling. However, adenomyosis was not seen in any uterus. ACTA2 immunostaining was less in the circular muscle layer of treated mice. The temporal pattern of desmin immunostaining found in control mice was absent in tamoxifen-treated mice. There was no difference in the localisation of laminin or fibronectin between control and tamoxifen-treated groups. However, laminin immunostaining was reduced in the circular muscle layer of treated mice. Vimentin could not be detected in either group. In conclusion, our results demonstrate that the development of the inner myometrium is particularly sensitive to oestrogen antagonism, and is affected by steroid receptor modulation. Although tamoxifen induces inner myometrial changes including that of ACTA2, desmin, ESR1 and laminin expression in C57/BL6J neonatal mice similar to those induced in CD-1 mice, C57/BL6J mice did not develop premature adenomyosis. Thus, disruption of the development and differentiation of the inner myometrium cannot alone explain the development of tamoxifen-associated adenomyosis, and this must be dependent upon its interaction with strain-dependent factors.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Endometriosis/etiología , Antagonistas de Estrógenos/administración & dosificación , Miometrio/efectos de los fármacos , Miometrio/crecimiento & desarrollo , Tamoxifeno/administración & dosificación , Actinas/análisis , Animales , Desmina/análisis , Receptor alfa de Estrógeno/análisis , Femenino , Fibronectinas/análisis , Inmunohistoquímica , Laminina/análisis , Ratones , Ratones Endogámicos C57BL , Miometrio/química , Vimentina/análisis
6.
Cells ; 9(6)2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32545544

RESUMEN

Early-life exposure of the myometrium to endocrine-disrupting chemicals (EDCs) has been shown to increase the risk of uterine fibroid (UF) prevalence in adulthood. Vitamin D3 (VitD3) is a unique, natural compound that may reduce the risk of developing UFs. However, little is known about the role and molecular mechanism of VitD3 on exposed myometrial stem cells (MMSCs). We investigated the role of, and molecular mechanism behind, VitD3 action on DNA damage response (DDR) defects in rat MMSCs due to developmental exposure to diethylstilbestrol (DES), with the additional goal of understanding how VitD3 decreases the incidence of UFs later in life. Female newborn Eker rats were exposed to DES or a vehicle early in life; they were then sacrificed at 5 months of age (pro-fibroid stage) and subjected to myometrial Stro1+/CD44+ stem cell isolation. Several techniques were performed to determine the effect of VitD3 treatment on the DNA repair pathway in DES-exposed MMSCs (DES-MMSCs). Results showed that there was a significantly reduced expression of RAD50 and MRE11, key DNA repair proteins in DES-exposed myometrial tissues, compared to vehicle (VEH)-exposed tissues (p < 0.01). VitD3 treatment significantly decreased the DNA damage levels in DES-MMSCs. Concomitantly, the levels of key DNA damage repair members, including the MRN complex, increased in DES-MMSCs following treatment with VitD3 (p < 0.01). VitD3 acts on DNA repair via the MRN complex/ATM axis, restores the DNA repair signaling network, and enhances DDR. This study demonstrates, for the first time, that VitD3 treatment attenuated the DNA damage load in MMSCs exposed to DES and classic DNA damage inducers. Moreover, VitD3 targets primed MMSCs, suggesting a novel therapeutic approach for the prevention of UF development.


Asunto(s)
Colecalciferol/farmacología , Daño del ADN/fisiología , Disruptores Endocrinos/metabolismo , Miometrio/metabolismo , Células Madre/citología , Animales , Colecalciferol/metabolismo , Disruptores Endocrinos/farmacología , Femenino , Leiomioma/metabolismo , Miometrio/crecimiento & desarrollo , Ratas
7.
Mol Cell Endocrinol ; 491: 110435, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31029737

RESUMEN

Proper myometrial adaptation during gestation is crucial for embryo implantation, pregnancy maintenance and parturition. Previously, we reported that neonatal exposure to endosulfan alters uterine development and induces implantation failures. The present work investigates the effects of endosulfan exposure on myometrial differentiation at the pre-implantation period, and myometrial activation during labor. Newborn female rats were s.c. injected with corn oil (vehicle) or 600 µg/kg/day of endosulfan (Endo600) on postnatal days (PND) 1, 3, 5 and 7. On PND90, the rats were mated to evaluate: i) the myometrial differentiation on gestational day 5 (GD5, pre-implantation period), by assessment myometrial histomorphology, smooth muscle cells (SMCs) proliferation, and expression of proteins involved in myometrial adaptation for embryo implantation (steroid receptors, Wnt7a and Hoxa10); ii) the timing of parturition and myometrial activation during labor by determining the uterine expression of contraction-associated genes (oxytocin receptor, OTXR; prostaglandin F2α receptor, PTGFR and connexin-43, Cx-43). Endosulfan decreased the thickness of both myometrial layers, with a concomitant decrease in the collagen remodeling. Blood vessels relative area in the interstitial connective tissue between muscle layers was also decreased. Endo600 group showed lower myometrial proliferation in association with a downregulation of Wnt7a and Hoxa10. Although in all females labor occurred on GD23, the exposure to endosulfan altered the timing of parturition, by inducing advancement in the initiation of labor. This alteration was associated with an increased uterine expression of OTXR, PTGFR and Cx-43. In conclusion, neonatal exposure to endosulfan produced long-term effects affecting myometrial adaptation during early pregnancy and labor. These alterations could be associated with the aberrant effects of endosulfan on the implantation process and the timing of parturition.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Endosulfano/administración & dosificación , Insecticidas/administración & dosificación , Trabajo de Parto/efectos de los fármacos , Miometrio/efectos de los fármacos , Contracción Uterina/efectos de los fármacos , Útero/efectos de los fármacos , Animales , Animales Recién Nacidos , Diferenciación Celular , Femenino , Trabajo de Parto/metabolismo , Miometrio/crecimiento & desarrollo , Miometrio/metabolismo , Embarazo , Ratas , Ratas Wistar , Contracción Uterina/metabolismo , Útero/crecimiento & desarrollo , Útero/metabolismo
8.
Reproduction ; 135(3): 357-65, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18299429

RESUMEN

Postnatal development of the uterus involves, particularly, development of uterine glands. Studies with ovariectomized ewe lambs demonstrated a role for ovaries in uterine growth and endometrial gland development between postnatal days (PNDs) 14 and 56. The uterotrophic ovarian factor(s) is presumably derived from the large numbers of growing follicles in the neonatal ovary present after PND 14. The Inverdale gene mutation (FecXI) results in an increased ovulation rate in heterozygous ewes; however, homozygous ewes (II) are infertile and have 'streak' ovaries that lack normal developing of preantral and antral follicles. Uteri were obtained on PND 56 to determine whether postnatal uterine development differs between wild-type (++) and II Inverdale ewes. When compared with wild-type ewes, uterine weight of II ewes was 52% lower, and uterine horn length tended to be shorter, resulting in a 68% reduction in uterine weight:length ratio in II ewes. Histomorphometrical analyses determined that endometria and myometria of II ewes were thinner and intercaruncular endometrium contained 38% fewer endometrial glands. Concentrations of estradiol in the neonatal ewes were low and not different between ++ and II ewes, but II ewes had lower concentrations of testosterone and inhibin-alpha between PNDs 14 and 56. Receptors for androgen and activin were detected in the neonatal uteri of both ++ and II ewes. These results support the concept that developing preantral and/or antral follicles of the ovary secrete uterotrophic factors, perhaps testosterone or inhibin-alpha, that acts in an endocrine manner to stimulate uterine growth and endometrial gland development in the neonatal ewes.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Infertilidad Femenina/fisiopatología , Ovario/crecimiento & desarrollo , Oveja Doméstica/crecimiento & desarrollo , Útero/crecimiento & desarrollo , Receptores de Activinas/análisis , Animales , Animales Recién Nacidos/genética , Cruzamiento , Endometrio/anatomía & histología , Endometrio/crecimiento & desarrollo , Estradiol/sangre , Femenino , Fertilidad/genética , Heterocigoto , Homocigoto , Inmunohistoquímica , Infertilidad Femenina/genética , Inhibinas/sangre , Miometrio/anatomía & histología , Miometrio/crecimiento & desarrollo , Ovariectomía , Ovario/química , Ovario/citología , Ovulación/genética , Receptores Androgénicos/análisis , Oveja Doméstica/genética , Testosterona/sangre , Útero/anatomía & histología , Útero/química
9.
Auton Neurosci ; 201: 32-39, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27639314

RESUMEN

Estrogen inhibits the growth and causes the degeneration (pruning) of sympathetic nerves supplying the rat myometrium. Previous cryoculture studies evidenced that substrate-bound signals contribute to diminish the ability of the estrogenized myometrium to support sympathetic nerve growth. Using electron microscopy, here we examined neurite-substrate interactions in myometrial cryocultures, observing that neurites grew associated to collagen fibrils present in the surface of the underlying cryosection. In addition, we assessed quantitatively the effects of estrogen on myometrial collagen organization in situ, using ovariectomized rats treated with estrogen and immature females undergoing puberty. Under low estrogen levels, most collagen fibrils were oriented in parallel to the muscle long axis (83% and 85%, respectively). Following estrogen treatment, 89% of fibrils was oriented perpendicularly to the muscle main axis; while after puberty, 57% of fibrils acquired this orientation. Immunohistochemistry combined with histology revealed that the vast majority of fine sympathetic nerve fibers supplying the myometrium courses within the areas where collagen realignment was observed. Finally, to assess whether depending on their orientation collagen fibrils can promote or inhibit neurite outgrowth, we employed cryocultures, now using as substrate tissue sections of rat-tail tendon. We observed that neurites grew extensively in the direction of the parallel-aligned collagen fibrils in the tendon main axis but were inhibited to grow perpendicularly to this axis. Collectively, these findings support the hypothesis that collagen reorientation may be one of the factors contributing to diminish the neuritogenic capacity of the estrogen-primed myometrial substrate.


Asunto(s)
Colágeno/metabolismo , Estrógenos/metabolismo , Miometrio/metabolismo , Animales , Técnicas de Cultivo de Célula , Colágeno/ultraestructura , Estrógenos/administración & dosificación , Femenino , Inmunohistoquímica , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Miometrio/citología , Miometrio/crecimiento & desarrollo , Miometrio/inervación , Proyección Neuronal/fisiología , Ovariectomía , Ratas Wistar , Maduración Sexual/fisiología , Simpatectomía , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/crecimiento & desarrollo , Sistema Nervioso Simpático/metabolismo , Cola (estructura animal)/metabolismo , Tendones/metabolismo
10.
J Pediatr Adolesc Gynecol ; 28(5): 387-94, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26248697

RESUMEN

STUDY OBJECTIVE: To evaluate myometrial growth and development. DESIGN: Thirty-five autopsy uteri, ranging from 10 weeks' gestation to age 18 years, acquired over 3 decades from 2 hospitals, were studied based on specimen availability, photographed for documentation, and reviewed at the end of the study. Most were embedded in toto, with 1 block and 1 slide per case. Some were immunostained for actin, CD10, MIB-1, and/or trichrome stain for collagen and muscle. Myometrial thickness was measured by ocular micrometry when sections were nontangential and analyzed by paired-sample t tests and bivariate linear regression. SETTING: Two university-affiliated hospitals. RESULTS: From 20 to 34 weeks, lateral wall corpus thickness increased 6-fold, with a 4- to 6-fold perinatal burst of growth (P < .01) and a drop in thickness after the neonatal period (P = .013). The corpus was thicker than the dome (P < .01) but less thick than the lower uterine segment (P = .087). The lower uterine segment was fully muscular in the second trimester, becoming more fibrous near term. Intramural, subserosal, and inframucosal myometaplasia were observed, as primitive stromal cells turned into muscle cells. Myometrial proliferation was brisk in the second trimester but greatly diminished in the perinatal period. Pressure effects from myometrial tone were observed during development. There was a pubertal burst of inframucosal myometaplasia. CONCLUSIONS: Myometaplasia accounted for most myometrial growth, especially in the perinatal and pubertal bursts of growth. Pressure effects, related to myometrial tone, appeared to affect myometrial development. True endocervix, with a fibrous wall and mucinous epithelium, appeared late in development.


Asunto(s)
Miometrio/crecimiento & desarrollo , Adolescente , Autopsia , Niño , Preescolar , Femenino , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Embarazo
11.
Steroids ; 68(1): 11-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12475719

RESUMEN

Although a clear role for estrogen receptor (ER) alpha has been established, the contribution of ERbeta in estrogen-dependent development, growth and functions of the myometrium is not understood. As a first step towards understanding the role of ERbeta, we have examined the expression of ERalpha and ERbeta in the human myometrium. With competitive RT-PCR assays, the level of ERbeta mRNA was 10-200 times lower than that of ERalpha mRNA in both premenopausal and postmenopausal myometrium. In premenopausal myometrium, the expression pattern of ERbeta mRNA during the menstrual cycle was similar to that of ERalpha mRNA, with highest levels in peri-ovulatory phase. In postmenopausal myometrium, ERbeta mRNA was significantly higher than it was in premenopausal myometrium, while the level of ERalpha mRNA was lower. The net result was a change in the ratio of ERbeta to ERalpha mRNA expression. The ratio changed from 0.6-1.5 in premenopausal to 2.5-7.6 in postmenopausal myometrium. In premenopausal women, the gonadotropin releasing hormone analogue, leuprorelin acetate, elicited a decrease in ERalpha and an increase in ERbeta mRNA expression to cause a postmenopausal receptor phenotype. Estradiol, on the other hand, reversed ERalpha and ERbeta mRNA expression and their ratio in postmenopausal myometrium to those of premenopausal myometrium. Immunohistochemical staining and Western blot analysis of ERalpha and ERbeta with semiquantitative analysis showed good agreement between mRNA and protein levels. The data indicate that coordinated expression of ERalpha and ERbeta might be necessary for normal estrogen action in myometrium. Furthermore, estrogen appears a dominant regulator of both receptors in the myometrium.


Asunto(s)
Miometrio/química , Posmenopausia/metabolismo , Premenopausia/metabolismo , Receptores de Estrógenos/análisis , Adulto , Western Blotting , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Humanos , Histerectomía , Inmunohistoquímica , Leuprolida/administración & dosificación , Leuprolida/farmacología , Ciclo Menstrual/metabolismo , Persona de Mediana Edad , Miometrio/efectos de los fármacos , Miometrio/crecimiento & desarrollo , ARN Mensajero/análisis , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Mol Endocrinol ; 28(3): 380-94, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24506537

RESUMEN

The smooth muscle layer of the uterus (ie, myometrium) is critical for a successful pregnancy and labor. We have shown that the conditional deletion of TGFß type 1 receptor (TGFBR1) in the female reproductive tract leads to remarkable smooth muscle defects. This study was aimed at defining the cellular and molecular basis of the myometrial defects. We found that TGFBR1 is required for myometrial configuration and formation during early postnatal uterine development. Despite the well-established role of TGFß signaling in vascular smooth muscle cell differentiation, the majority of smooth muscle genes were expressed in Tgfbr1 conditional knockout (cKO) uteri at similar levels as controls during postnatal uterine development, coinciding with the presence but abnormal distribution of proteins for select smooth muscle markers. Importantly, the uteri of these mice had impaired synthesis of key extracellular matrix proteins and dysregulated expression of platelet-derived growth factors. Furthermore, platelet-derived growth factors induced the migration of uterine stromal cells from both control and Tgfbr1 cKO mice in vitro. Our results suggest that the myometrial defects in Tgfbr1 cKO mice may not directly arise from an intrinsic deficiency in uterine smooth muscle cell differentiation but are linked to the impaired production of key extracellular matrix components and abnormal uterine cell migration during a critical time window of postnatal uterine development. These findings will potentially aid in the design of novel therapies for reproductive disorders associated with myometrial defects.


Asunto(s)
Miometrio/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Animales , Movimiento Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Expresión Génica , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/fisiología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Células del Estroma/fisiología , Factor de Crecimiento Transformador beta/fisiología
13.
Mol Endocrinol ; 27(9): 1442-54, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23904126

RESUMEN

Müllerian duct differentiation and development into the female reproductive tract is essential for fertility, but mechanisms regulating these processes are poorly understood. WNT signaling is critical for proper development of the female reproductive tract as evident by the phenotypes of Wnt4, Wnt5a, Wnt7a, and ß-catenin (Ctnnb1) mutant mice. Here we extend these findings by determining the effects of constitutive CTNNB1 activation within the mesenchyme of the developing Müllerian duct and its differentiated derivatives. This was accomplished by crossing Amhr2-Cre knock-in mice with Ctnnb1 exon (ex) 3(f/f) mice. Amhr2-Cre(Δ/+); Ctnnb1 ex3(f/+) females did not form an oviduct, had smaller uteri, endometrial gland defects, and were infertile. At the cellular level, stabilization of CTNNB1 in the mesenchyme caused alterations within the epithelium, including less proliferation, delayed uterine gland formation, and induction of an epithelial-mesenchymal transition (EMT) event. This EMT event is observed before birth and is complete within 5 days after birth. Misexpression of estrogen receptor α in the epithelia correlated with the EMT before birth, but not after. These studies indicate that regulated CTNNB1 in mesenchyme is important for epithelial cell differentiation during female reproductive tract development.


Asunto(s)
Diferenciación Celular , Células Epiteliales/patología , Mesodermo/metabolismo , Conductos Paramesonéfricos/crecimiento & desarrollo , Conductos Paramesonéfricos/metabolismo , Útero/crecimiento & desarrollo , Útero/metabolismo , beta Catenina/metabolismo , Animales , Animales Recién Nacidos , Neoplasias Endometriales/patología , Endometrio/anomalías , Endometrio/crecimiento & desarrollo , Endometrio/patología , Transición Epitelial-Mesenquimal , Epitelio/crecimiento & desarrollo , Epitelio/patología , Femenino , Infertilidad Femenina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Conductos Paramesonéfricos/patología , Miometrio/anomalías , Miometrio/crecimiento & desarrollo , Miometrio/patología , Estabilidad Proteica , Sarcoma/patología , Células del Estroma/patología , Útero/patología
14.
Reprod Sci ; 17(9): 791-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20601542

RESUMEN

L-type amino acid transporter 1 (LAT1) and LAT2 were shown to encode system L, which mediates the Na(+)-independent transport of branched-chain and aromatic amino acids. We demonstrated previously that LAT2 is a progesterone receptor target gene involved in leiomyoma growth. The role of LAT1 in the regulation of human uterine leiomyoma growth, however, remains unelucidated. We herein investigated the function of LAT1 and its progesterone-mediated regulation within human uterine leiomyoma smooth muscle (LSM) cells (n = 8) and tissues (n = 29). In vivo, LAT1 expression was higher in leiomyoma than in myometrial tissue. LAT1 knockdown augmented cell proliferation and viability. Treatment of LSM cells with RU486 markedly increased LAT1 messenger RNA (mRNA) levels but decreased proliferation in a dose-dependent manner. L-type amino acid transporter 1 as a downstream target, however, did not entirely account for this antiproliferative effect of RU486 on LSM cells. Taken together, LAT1 may have a critical and complex role in regulating human leiomyoma cell growth.


Asunto(s)
Proliferación Celular , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Leiomioma/metabolismo , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas de Hormonas/farmacología , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , Leiomioma/genética , Leiomioma/patología , Persona de Mediana Edad , Mifepristona/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Miometrio/efectos de los fármacos , Miometrio/crecimiento & desarrollo , Miometrio/patología , Progesterona/antagonistas & inhibidores , Progesterona/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
16.
Endocrinology ; 150(10): 4672-80, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19589861

RESUMEN

The adaptive growth of the uterus during gestation involves gradual changes in cellular phenotypes from the early proliferative to the intermediate synthetic phase of cellular hypertrophy, ending in the final contractile/labour phenotype. The mammalian target of rapamycin (mTOR) signaling pathway regulates cell growth and proliferation in many tissues. We hypothesized that mTOR was a mediator of hormone-initiated myometrial hyperplasia during gestation. The protein expression and phosphorylation levels of mTOR, its upstream regulators [insulin receptor substrate-1, phosphoinositide-3-kinase (PI3K), Akt], and downstream effectors [S6-kinase-1 (S6K1) and eI4FE-binding protein 1 (4EBP1)] were analyzed throughout normal pregnancy in rats. In addition, we used an ovariectomized (OVX) rat model to analyze the modulation of the mTOR pathway and proliferative activity of the uterine myocytes by estradiol alone and in combination with the mTOR-specific inhibitor rapamycin. Our results demonstrate that insulin receptor substrate-1 protein levels and the phosphorylated (activated) forms of PI3K, mTOR, and S6K1 were significantly up-regulated in the rat myometrium during the proliferative phase of pregnancy. Treatment of the OVX rats with estradiol caused a transient increase in IGF-I followed by an up-regulation of the PI3K/mTOR pathway, which became apparent by a cascade of phosphorylation reactions (P-P85, P-Akt, P-mTOR, P-S6K1, and P-4EBP1). Rapamycin blocked activation of P-mTOR, P-S6K1, and P-4EBP1 proteins and significantly reduced the number of proliferating cells in the myometrium of OVX rats. Our in vivo data demonstrate that estradiol was able to activate the PI3K/mTOR signaling pathway in uterine myocytes and suggest that this activation is responsible for the induction of myometrial hyperplasia during early gestation.


Asunto(s)
Proliferación Celular , Estradiol/metabolismo , Miometrio/crecimiento & desarrollo , Embarazo/fisiología , Proteínas Quinasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Activación Enzimática , Femenino , Expresión Génica , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Miocitos del Músculo Liso/fisiología , Miometrio/metabolismo , Ovariectomía , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Sirolimus , Serina-Treonina Quinasas TOR
17.
J Cell Mol Med ; 12(4): 1360-73, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18363833

RESUMEN

The smooth muscle of the uterus during pregnancy presents a unique circumstance of physiological mechanotransduction as the tissue remodels in response to stretches imposed by the growing foetus(es), yet the nature of the molecular and functional adaptations remain unresolved. We studied, in myometrium isolated from non-pregnant (NP) and pregnant mice, the active and passive length-tension curves by myography and the expression and activation by immunoblotting of focal adhesion-related proteins known in other systems to participate in mechanosensing and mechanotransduction. In situ uterine mass correlated with pup number and weight throughout pregnancy. In vitro myometrial active, and passive, length-tension curves shifted significantly to the right during pregnancy indicative of altered mechanosensitivity; at term, maximum active tension was generated following 3.94+/-0.33-fold stretch beyond slack length compared to 1.91+/-0.12-fold for NP mice. Moreover, mechanotransduction was altered during pregnancy as evidenced by the progressive increase in absolute force production at each optimal stretch. Pregnancy was concomitantly associated with an increased expression of the dense plaque-associated proteins FAK and paxillin, and elevated activation of FAK, paxillin, c-Src and extracellular signal-regulated kinase (ERK1/2) which reversed 1 day post-partum. Electron microscopy revealed close appositioning of neighbouring myometrial cells across a narrow extracellular cleft adjoining plasmalemmal dense plaques. Collectively, these results suggest a physiological basis of myometrial length adaptation, long known to be a property of many smooth muscles, whereupon plasmalemmal dense plaque proteins serve as molecular signalling and structural platforms contributing to functional (contractile) remodelling in response to chronic stretch.


Asunto(s)
Adaptación Fisiológica , Miometrio/fisiología , Preñez/fisiología , Actinas/metabolismo , Animales , Fenómenos Biomecánicos , Peso Corporal , Ciclo Estral/fisiología , Femenino , Ratones , Modelos Biológicos , Miometrio/crecimiento & desarrollo , Miometrio/ultraestructura , Tamaño de los Órganos , Embarazo , Proteínas Gestacionales/metabolismo , Útero/crecimiento & desarrollo
18.
Biol Reprod ; 76(4): 571-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17123939

RESUMEN

While the insulin-like growth factor (IGF) system is known to regulate uterine function during the estrous cycle, there are limited data on its role in myometrial growth and development during pregnancy. To address this issue, we defined the expression of the Igf hormones (1 and 2), their binding proteins (Igfbp 1-6), and Igf1r receptor genes in pregnant, laboring, and postpartum rat myometrium by real-time PCR. IGF family genes were differentially expressed throughout gestation. Igf1 and Igfbp1 mRNA levels were upregulated during proliferative phase (Days 6-12) of rat gestation. Igfbp3 gene expression also was elevated in proliferating smooth muscle cells (SMCs) and was highest at the time of transition between proliferative and synthetic phases (Days 12-15). Igfbp6 gene expression profile paralleled plasma progesterone (P4) concentrations, peaking during the synthetic phase (Days 17-19) and decreasing thereafter. Administration of P4 at late pregnancy (starting from Day 20) to maintain elevated plasma P4 concentrations blocked the onset of labor and prevented the fall in Igfbp6 mRNA levels. In contrast, the treatment of pregnant rats with the P4 receptor antagonist RU486 on Day 19 induced preterm labor and the premature decrease of Igfbp6 gene expression. Igfbp2 gene expression was transiently upregulated during the contractile phase of gestation (Days 21-23) solely in the gravid horn of unilaterally pregnant rats, but it was not affected in P4- or RU486-treated animals, supporting a role for mechanical stretch imposed by the growing fetuses. Igfbp5 gene was induced during postpartum involution. Our results suggest the importance of the IGF system in phenotypic and functional changes of myometrial SMCs throughout gestation in preparation for labor.


Asunto(s)
Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Desarrollo de Músculos/fisiología , Miometrio/crecimiento & desarrollo , Preñez , Somatomedinas/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Número de Embarazos/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Modelos Biológicos , Desarrollo de Músculos/genética , Miometrio/metabolismo , Periodo Posparto/genética , Periodo Posparto/metabolismo , Embarazo , Preñez/genética , Preñez/metabolismo , Progesterona/farmacología , Ratas , Ratas Wistar , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Somatomedinas/genética , Somatomedinas/metabolismo
19.
Reprod Sci ; 14(2): 121-36, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17636224

RESUMEN

Epidemiological and experimental animal studies have shown that exposure to xenoestrogens during reproductive tract development reprograms target tissues, leading to increased disease risk later in adult life. To understand what defines the critical risk period for this effect, termed developmental programming, the authors assess the sensitivity of the female reproductive tract to developmental programming during various stages of neonatal development. Eker rats, which are predisposed to develop uterine leiomyoma because of a germ-line defect in the tuberous sclerosis complex 2 (Tsc-2) tumor suppressor gene, were exposed to the xenoestrogen diethylstilbestrol (DES) on either postnatal days 3 to 5, 10 to 12, or 17 to 19, 3 important periods of reproductive tract development and differentiation. Developmental programming was observed in both carrier (Tsc-2(Ek/+)) and wild-type (Tsc-2(+/+)) rats exposed to DES at days 3 to 5 and days 10 to 12 but not in rats exposed at days 17 to 19. Developmental programming resulted in increased tumor suppressor gene penetrance in Tsc-2(Ek/+) females relative to vehicle controls. In contrast, DES exposure at days 17 to 19 did not significantly increase the incidence of uterine leiomyoma in carrier females, indicating that the window of susceptibility had closed by this time. Gene expression analysis to determine what defined the susceptible (days 3-5 and days 10-12) versus resistant (days 17-19) periods revealed that in adult myometrium, expression of the estrogen-responsive genes calbindin D(9)K and progesterone receptor had been reprogrammed in females exposed to DES at days 3 to 5 and days 10 to 12 but not in those exposed at days 17 to 19. Reprogramming in response to DES exposure resulted in a hyperresponsiveness to ovarian hormones and could be prevented by ovariectomy prior to sexual maturity. Furthermore, in the neonatal uterus, DES was equally effective at inducing transcription of estrogen-responsive genes during both sensitive and resistant periods, indicating that resistance to developmental programming was not due to an inability of the estrogen receptor to transactivate gene expression. Interestingly, the resistant period coincided with the time at which reproductive tract tissues are exposed to endogenous estrogen, suggesting that target tissues are most vulnerable to developmental programming during the period in which they would normally be maintained in an estrogen-naïve state.


Asunto(s)
Dietilestilbestrol/toxicidad , Estrógenos no Esteroides/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Leiomioma/inducido químicamente , Proteínas Supresoras de Tumor/genética , Neoplasias Uterinas/inducido químicamente , Útero/efectos de los fármacos , Animales , Calbindinas , Dietilestilbestrol/administración & dosificación , Modelos Animales de Enfermedad , Estrógenos/fisiología , Estrógenos no Esteroides/administración & dosificación , Femenino , Predisposición Genética a la Enfermedad , Immunoblotting , Leiomioma/genética , Leiomioma/fisiopatología , Ciclo Menstrual/fisiología , Miometrio/efectos de los fármacos , Miometrio/crecimiento & desarrollo , Miometrio/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Ratas Mutantes , Receptores de Progesterona/genética , Proteína G de Unión al Calcio S100/genética , Factores de Tiempo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Neoplasias Uterinas/genética , Neoplasias Uterinas/fisiopatología , Útero/crecimiento & desarrollo , Útero/metabolismo
20.
Dev Biol ; 288(1): 276-83, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16256976

RESUMEN

Precise cell fate decisions during differentiation of uterine tissues from the embryonic Müllerian duct are critical for normal fertility. Wnt-7a, a member of the Wnt family of secreted signaling molecules that can signal through a canonical beta-catenin pathway, is necessary for the correct differentiation of both anterior/posterior and radial axes of the uterus. In order to investigate the role of beta-catenin directly in mouse uterine development, we have generated mice that are deficient in beta-catenin expression in the embryonic Müllerian duct. We have found that conditional deletion of beta-catenin in the Müllerian duct mesenchyme before postnatal differentiation of the uterine layers results in a phenotype that is distinct from the phenotype observed by deletion of Wnt-7a. Shortly after birth, the uteri of the conditional mutants appear smaller and less organized. The uteri of adult conditional beta-catenin mutants are grossly deficient in smooth muscle of the myometrium, which has been replaced by adipose, a phenotype resembling human lipoleiomyoma. We also show that the adipocytes in the uteri of mice conditionally deleted for beta-catenin are derived from Müllerian inhibiting substance type II receptor-expressing cells suggesting that they share a common origin with the uterine smooth muscle cells. These results describe the first molecular evidence linking disruption of beta-catenin expression in mesenchymal cells with a switch from myogenesis to adipogenesis in vivo.


Asunto(s)
Adipogénesis/genética , Eliminación de Gen , Mesodermo/fisiología , Miometrio/embriología , Útero/embriología , beta Catenina/deficiencia , Adipocitos/fisiología , Tejido Adiposo/fisiología , Animales , Femenino , Integrasas/biosíntesis , Integrasas/genética , Ratones , Ratones Transgénicos , Músculo Liso/metabolismo , Miometrio/crecimiento & desarrollo , Útero/crecimiento & desarrollo , beta Catenina/genética , beta Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA