Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Agric Food Chem ; 69(5): 1693-1704, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33528250

ABSTRACT

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.


Subject(s)
Genistein/pharmacology , Phytoestrogens/pharmacology , Uterus/drug effects , Uterus/growth & development , Animals , Cell Proliferation/drug effects , Dietary Exposure , Female , Mice , Myometrium/drug effects , Myometrium/growth & development
2.
Cells ; 9(6)2020 06 12.
Article in English | MEDLINE | ID: mdl-32545544

ABSTRACT

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.


Subject(s)
Cholecalciferol/pharmacology , DNA Damage/physiology , Endocrine Disruptors/metabolism , Myometrium/metabolism , Stem Cells/cytology , Animals , Cholecalciferol/metabolism , Endocrine Disruptors/pharmacology , Female , Leiomyoma/metabolism , Myometrium/growth & development , Rats
3.
Mol Cell Endocrinol ; 491: 110435, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31029737

ABSTRACT

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.


Subject(s)
Adaptation, Physiological/drug effects , Endosulfan/administration & dosage , Insecticides/administration & dosage , Labor, Obstetric/drug effects , Myometrium/drug effects , Uterine Contraction/drug effects , Uterus/drug effects , Animals , Animals, Newborn , Cell Differentiation , Female , Labor, Obstetric/metabolism , Myometrium/growth & development , Myometrium/metabolism , Pregnancy , Rats , Rats, Wistar , Uterine Contraction/metabolism , Uterus/growth & development , Uterus/metabolism
4.
Biol Reprod ; 99(4): 735-748, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29688260

ABSTRACT

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.


Subject(s)
DNA Repair/drug effects , Endocrine Disruptors/toxicity , Leiomyoma/etiology , Myometrium/drug effects , Myometrium/metabolism , Uterine Neoplasms/etiology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , DNA Damage , DNA Repair/genetics , Diethylstilbestrol/toxicity , Disease Models, Animal , Female , Germ-Line Mutation , Humans , Leiomyoma/genetics , Leiomyoma/metabolism , Mediator Complex/genetics , Myometrium/growth & development , Rats , Rats, Mutant Strains , Tuberous Sclerosis Complex 2 Protein/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
5.
Auton Neurosci ; 201: 32-39, 2016 12.
Article in English | MEDLINE | ID: mdl-27639314

ABSTRACT

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.


Subject(s)
Collagen/metabolism , Estrogens/metabolism , Myometrium/metabolism , Animals , Cell Culture Techniques , Collagen/ultrastructure , Estrogens/administration & dosage , Female , Immunohistochemistry , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Myometrium/growth & development , Myometrium/innervation , Neuronal Outgrowth/physiology , Ovariectomy , Rats, Wistar , Sexual Maturation/physiology , Sympathectomy , Sympathetic Nervous System/cytology , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/metabolism , Tail/metabolism , Tendons/metabolism
6.
J Pediatr Adolesc Gynecol ; 28(5): 387-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26248697

ABSTRACT

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.


Subject(s)
Myometrium/growth & development , Adolescent , Autopsy , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Infant, Newborn , Pregnancy
7.
Reprod Fertil Dev ; 27(7): 985-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24630125

ABSTRACT

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.


Subject(s)
Follistatin/genetics , Oviducts/growth & development , Uterus/growth & development , Animals , Cell Proliferation/genetics , Endometrium/growth & development , Endometrium/metabolism , Estrogens/pharmacology , Female , Follistatin/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mice, Transgenic , Myometrium/growth & development , Myometrium/metabolism , Ovariectomy , Oviducts/diagnostic imaging , Oviducts/metabolism , Uterus/drug effects , Uterus/metabolism
8.
Mol Endocrinol ; 28(3): 380-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24506537

ABSTRACT

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.


Subject(s)
Myometrium/growth & development , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Animals , Cell Movement , Cells, Cultured , Extracellular Matrix/metabolism , Female , Gene Expression , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/physiology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Receptor, Transforming Growth Factor-beta Type I , Stromal Cells/physiology , Transforming Growth Factor beta/physiology
9.
Mol Endocrinol ; 27(9): 1442-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23904126

ABSTRACT

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.


Subject(s)
Cell Differentiation , Epithelial Cells/pathology , Mesoderm/metabolism , Mullerian Ducts/growth & development , Mullerian Ducts/metabolism , Uterus/growth & development , Uterus/metabolism , beta Catenin/metabolism , Animals , Animals, Newborn , Endometrial Neoplasms/pathology , Endometrium/abnormalities , Endometrium/growth & development , Endometrium/pathology , Epithelial-Mesenchymal Transition , Epithelium/growth & development , Epithelium/pathology , Female , Infertility, Female/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mullerian Ducts/pathology , Myometrium/abnormalities , Myometrium/growth & development , Myometrium/pathology , Protein Stability , Sarcoma/pathology , Stromal Cells/pathology , Uterus/pathology
10.
PLoS Genet ; 7(8): e1002210, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21852953

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Myometrium/growth & development , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Animals , Body Patterning , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Female , Gene Expression Regulation, Developmental , Hermaphroditic Organisms , Mutation , Myometrium/metabolism , Phenotype , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Two-Hybrid System Techniques
11.
Reprod Sci ; 17(9): 791-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20601542

ABSTRACT

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.


Subject(s)
Cell Proliferation , Large Neutral Amino Acid-Transporter 1/metabolism , Leiomyoma/metabolism , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Uterine Neoplasms/metabolism , Adult , Cell Proliferation/drug effects , Cell Survival , Dose-Response Relationship, Drug , Female , Hormone Antagonists/pharmacology , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leiomyoma/genetics , Leiomyoma/pathology , Middle Aged , Mifepristone/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myometrium/drug effects , Myometrium/growth & development , Myometrium/pathology , Progesterone/antagonists & inhibitors , Progesterone/metabolism , RNA Interference , RNA, Messenger/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
12.
Reproduction ; 139(6): 1067-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20368191

ABSTRACT

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.


Subject(s)
Animals, Newborn/growth & development , Endometriosis/etiology , Estrogen Antagonists/administration & dosage , Myometrium/drug effects , Myometrium/growth & development , Tamoxifen/administration & dosage , Actins/analysis , Animals , Desmin/analysis , Estrogen Receptor alpha/analysis , Female , Fibronectins/analysis , Immunohistochemistry , Laminin/analysis , Mice , Mice, Inbred C57BL , Myometrium/chemistry , Vimentin/analysis
13.
Endocrinology ; 150(10): 4672-80, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19589861

ABSTRACT

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.


Subject(s)
Cell Proliferation , Estradiol/metabolism , Myometrium/growth & development , Pregnancy/physiology , Protein Kinases/metabolism , Animals , Carrier Proteins/metabolism , Enzyme Activation , Female , Gene Expression , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins , Myocytes, Smooth Muscle/physiology , Myometrium/metabolism , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases
14.
J Cell Mol Med ; 12(4): 1360-73, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18363833

ABSTRACT

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.


Subject(s)
Adaptation, Physiological , Myometrium/physiology , Pregnancy, Animal/physiology , Actins/metabolism , Animals , Biomechanical Phenomena , Body Weight , Estrous Cycle/physiology , Female , Mice , Models, Biological , Myometrium/growth & development , Myometrium/ultrastructure , Organ Size , Pregnancy , Pregnancy Proteins/metabolism , Uterus/growth & development
15.
Reproduction ; 135(3): 357-65, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18299429

ABSTRACT

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.


Subject(s)
Animals, Newborn/growth & development , Infertility, Female/physiopathology , Ovary/growth & development , Sheep, Domestic/growth & development , Uterus/growth & development , Activin Receptors/analysis , Animals , Animals, Newborn/genetics , Breeding , Endometrium/anatomy & histology , Endometrium/growth & development , Estradiol/blood , Female , Fertility/genetics , Heterozygote , Homozygote , Immunohistochemistry , Infertility, Female/genetics , Inhibins/blood , Myometrium/anatomy & histology , Myometrium/growth & development , Ovariectomy , Ovary/chemistry , Ovary/cytology , Ovulation/genetics , Receptors, Androgen/analysis , Sheep, Domestic/genetics , Testosterone/blood , Uterus/anatomy & histology , Uterus/chemistry
16.
Reprod Sci ; 14(2): 121-36, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17636224

ABSTRACT

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.


Subject(s)
Diethylstilbestrol/toxicity , Estrogens, Non-Steroidal/toxicity , Gene Expression Regulation, Developmental/drug effects , Leiomyoma/chemically induced , Tumor Suppressor Proteins/genetics , Uterine Neoplasms/chemically induced , Uterus/drug effects , Animals , Calbindins , Diethylstilbestrol/administration & dosage , Disease Models, Animal , Estrogens/physiology , Estrogens, Non-Steroidal/administration & dosage , Female , Genetic Predisposition to Disease , Immunoblotting , Leiomyoma/genetics , Leiomyoma/physiopathology , Menstrual Cycle/physiology , Myometrium/drug effects , Myometrium/growth & development , Myometrium/metabolism , Polymerase Chain Reaction , Rats , Rats, Mutant Strains , Receptors, Progesterone/genetics , S100 Calcium Binding Protein G/genetics , Time Factors , Tuberous Sclerosis Complex 2 Protein , Uterine Neoplasms/genetics , Uterine Neoplasms/physiopathology , Uterus/growth & development , Uterus/metabolism
17.
Biol Reprod ; 76(4): 571-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17123939

ABSTRACT

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.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/physiology , Muscle Development/physiology , Myometrium/growth & development , Pregnancy, Animal , Somatomedins/physiology , Animals , Female , Gene Expression Regulation, Developmental/drug effects , Gravidity/physiology , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Male , Models, Biological , Muscle Development/genetics , Myometrium/metabolism , Postpartum Period/genetics , Postpartum Period/metabolism , Pregnancy , Pregnancy, Animal/genetics , Pregnancy, Animal/metabolism , Progesterone/pharmacology , Rats , Rats, Wistar , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Somatomedins/genetics , Somatomedins/metabolism
18.
Dev Biol ; 288(1): 276-83, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16256976

ABSTRACT

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.


Subject(s)
Adipogenesis/genetics , Gene Deletion , Mesoderm/physiology , Myometrium/embryology , Uterus/embryology , beta Catenin/deficiency , Adipocytes/physiology , Adipose Tissue/physiology , Animals , Female , Integrases/biosynthesis , Integrases/genetics , Mice , Mice, Transgenic , Muscle, Smooth/metabolism , Myometrium/growth & development , Uterus/growth & development , beta Catenin/genetics , beta Catenin/physiology
19.
Mol Hum Reprod ; 11(8): 561-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051682

ABSTRACT

Uterine leiomyomas (fibroids) are the most frequent tumour of the female reproductive tract and are the primary cause of hysterectomies in women worldwide. Effective treatment options are few. In a search for alternative treatments, we have established primary cultures of human leiomyoma cells and adjacent myometrial tissues, and documented their growth dynamics in response to estradiol (E2) and pioglitazone (PIO), a peroxisome proliferation-activated receptor-gamma (PPARgamma) ligand, currently in clinical use for type II diabetes mellitus. Human uterine primary cell cultures display morphology and desmin content consistent with their smooth muscle origin. Surprisingly, leiomyoma cells exhibited slower proliferation patterns relative to matched myometrial cells, both in the absence and presence of E2, suggesting that tumour genesis may not be because of increased growth potential but could be related to suppression of growth-inhibiting factors in vivo. PIO significantly inhibited the cell proliferation of both myometrial and leiomyoma cells in a dose-dependent manner. Our results suggest the possibility of using PPARgamma ligands, such as PIO, as therapeutic agents for the conservative management of uterine fibroids.


Subject(s)
Leiomyoma/drug therapy , Leiomyoma/pathology , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Adult , Cell Division/drug effects , Cell Division/physiology , Female , Humans , Ligands , Middle Aged , Muscle Development/physiology , Myometrium/cytology , Myometrium/growth & development , Pioglitazone , Tumor Cells, Cultured
20.
Biol Reprod ; 69(3): 843-50, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12748120

ABSTRACT

Uterine gland development or adenogenesis in the neonatal ovine uterus involves budding and tubulogenesis followed by coiling and branching morphogenesis of the glandular epithelium (GE) from the luminal epithelium (LE) between birth (Postnatal Day [PND] 0) and PND 56. Activins, which are members of the transforming growth factor beta superfamily, and follistatin, an inhibitor of activins, regulate epithelial branching morphogenesis in other organs. The objective of the present study was to determine effects of postnatal age on expression of follistatin, inhibin alpha subunit, betaA subunit, betaB subunit, activin receptor (ActR) type IA, ActRIB, and ActRII in the developing ovine uterus. Ewes were ovariohysterectomized on PND 0, 7, 14, 21, 28, 35, 42, 49, or 56. The uterus was analyzed by in situ hybridization and immunohistochemistry. Neither inhibin alpha subunit mRNA or protein was detected in the neonatal uterus. Expression of betaA and betaB subunits was detected predominantly in the endometrial LE and GE and myometrium between PND 0 and PND 56. In all uterine cell types, ActRIA, ActRIB, and ActRII were expressed, with the highest levels observed in the endometrial LE and GE and myometrium. Between PND 0 and PND 14, follistatin was detected in all uterine cell types. However, between PND 21 and PND 56, follistatin was only detected in the stroma and myometrium and not in the developing GE. Collectively, the present results indicate that components of the activin-follistatin system are expressed in the developing neonatal ovine uterus and are potential regulators of endometrial gland morphogenesis.


Subject(s)
Activins/metabolism , Animals, Newborn/physiology , Endometrium/growth & development , Epithelium/growth & development , Follistatin/metabolism , Sheep/physiology , Activin Receptors/genetics , Activin Receptors/metabolism , Animals , Cell Differentiation , Cell Division , Endometrium/cytology , Endometrium/metabolism , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Inhibins/genetics , Inhibins/metabolism , Morphogenesis/physiology , Myometrium/cytology , Myometrium/growth & development , Myometrium/metabolism , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...