Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 96(1): 1, 2017 01 01.
Article in English | MEDLINE | ID: mdl-29554246
4.
Biol Reprod ; 90(1): 7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227756

ABSTRACT

Fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate luminal epithelial (LE) cell proliferation in the adult mouse uterus. This study tested the hypothesis that FGFR2 has a biological role in postnatal development and function of the uterus by conditionally deleting Fgfr2 after birth using progesterone receptor (Pgr)-Cre mice. Adult Fgfr2 mutant female mice were initially subfertile and became infertile with increasing parity. No defects in uterine gland development were observed in conditional Fgfr2 mutant mice. In the adult, Fgfr2 mutant mice possessed a histologically normal reproductive tract with the exception of the uterus. The LE of the Fgfr2 mutant uterus was stratified, but no obvious histological differences were observed in the glandular epithelium, stroma, or myometrium. Within the stratified LE, cuboidal basal cells were present and positive for basal cell markers (KRT14 and TRP63). Nulliparous bred Fgfr2 mutants contained normal numbers of blastocysts on Day 3.5 postmating, but the number of embryo implantation sites was substantially reduced on Day 5.5 postmating. These results support the idea that loss of FGFR2 in the uterus after birth alters its development, resulting in LE stratification and peri-implantation pregnancy loss.


Subject(s)
Epithelial Cells/physiology , Fertility/genetics , Receptor, Fibroblast Growth Factor, Type 2/physiology , Uterus/cytology , Uterus/physiology , Animals , Embryo Implantation/genetics , Embryo Loss/genetics , Female , Genotype , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Uterus/growth & development
5.
Endocrinology ; 154(1): 446-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142810

ABSTRACT

Differentiation of endometrial stromal cells into decidual cells is a prerequisite for successful embryo implantation. Our previous studies in the mouse have shown that bone morphogenetic protein 2 (BMP2), a morphogen belonging to the TGFß superfamily, is essential for this differentiation process. BMP2 is markedly induced in human primary endometrial stromal cells (HESCs) as they undergo differentiation in response to steroid hormones and cAMP. The present study was undertaken to identify the BMP2-mediated molecular pathways in primary cultures of HESCs during decidualization. Using gene expression profiling, we identified wingless-related murine mammary tumor virus integration site 4 (WNT4) as a target of BMP2 regulation during decidualization. Attenuation of WNT4 expression in HESCs by small interfering RNA administration greatly reduced BMP2-induced stromal differentiation. Additionally, adenovirus-mediated overexpression of WNT4 in HESCs markedly advanced the differentiation program, indicating that it is a key regulator of decidualization. The stimulatory effect of WNT4 was accompanied by the accumulation of active ß-catenin in the nuclei of decidualizing stromal cells, indicating the involvement of the canonical WNT signaling pathway. Functional inhibition of WNT4/ß-catenin pathway by Dickkopf-1, an inhibitor of the canonical WNT signaling, or small interfering RNA-mediated silencing of ß-catenin expression, greatly reduced the BMP2- and WNT4-induced decidualization. Gene expression profiling revealed that Forkhead box protein O1, a forkhead family transcription factor and previously reported regulator of HESC differentiation, is a common downstream mediator of both BMP2 and WNT4 signaling. Taken together, these studies uncovered a linear pathway involving BMP2, WNT4/ß-catenin, and Forkhead box protein O1 that operates in human endometrium to critically control decidualization.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/physiology , Endometrium/cytology , Stromal Cells/cytology , Stromal Cells/metabolism , Wnt4 Protein/metabolism , beta Catenin/metabolism , Bone Morphogenetic Protein 2/genetics , Cell Differentiation/genetics , Cells, Cultured , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , RNA, Small Interfering , Signal Transduction/genetics , Signal Transduction/physiology , Wnt4 Protein/genetics , beta Catenin/genetics
6.
FASEB J ; 26(1): 282-94, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21990372

ABSTRACT

Uterine receptivity implies a dialogue between the hormonally primed maternal endometrium and the free-floating blastocyst. Endometrial stromal cells proliferate, avert apoptosis, and undergo decidualization in preparation for implantation; however, the molecular mechanisms that underlie differentiation into the decidual phenotype remain largely undefined. The Notch family of transmembrane receptors transduce extracellular signals responsible for cell survival, cell-to-cell communication, and differentiation, all fundamental processes for decidualization and pregnancy. Using a murine artificial decidualization model, pharmacological inhibition of Notch signaling by γ-secretase inhibition resulted in a significantly decreased deciduoma. Furthermore, a progesterone receptor (PR)-Cre Notch1 bigenic (Notch1(d/d)) confirmed a Notch1-dependent hypomorphic decidual phenotype. Microarray and pathway analysis, following Notch1 ablation, demonstrated significantly altered signaling repertoire. Concomitantly, hierarchical clustering demonstrated Notch1-dependent differences in gene expression. Uteri deprived of Notch1 signaling demonstrated decreased cellular proliferation; namely, reduced proliferation-specific antigen, Ki67, altered p21, cdk6, and cyclinD activity and an increased apoptotic-profile, cleaved caspase-3, Bad, and attenuated Bcl2. The results demonstrate that the preimplantation uterus relies on Notch signaling to inhibit apoptosis of stromal fibroblasts and regulate cell cycle progression, which together promotes successful decidualization. In summary, Notch1 signaling modulates multiple signaling mechanisms crucial for decidualization and these studies provide additional perspectives to the coordination of multiple signaling modalities required during decidualization.


Subject(s)
Decidua/physiology , Embryo Implantation/physiology , Pregnancy, Animal/physiology , Receptor, Notch1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Animals , Apoptosis/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cytoskeleton/metabolism , Decidua/cytology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Ovariectomy , Pregnancy , Receptor, Notch1/genetics , Signal Transduction/physiology
7.
Endocrinology ; 152(11): 4336-49, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21862609

ABSTRACT

Estrogen receptor (ER) is a key regulator of mammary gland development and is also implicated in breast tumorigenesis. Because ER-mediated activities depend critically on coregulator partner proteins, we have investigated the consequences of reduction or loss of function of the coregulator repressor of ER activity (REA) by conditionally deleting one allele or both alleles of the REA gene at different stages of mammary gland development. Notably, we find that heterozygosity and nullizygosity for REA result in very different mammary phenotypes and that REA has essential roles in the distinct morphogenesis and functions of the mammary gland at different stages of development, pregnancy, and lactation. During puberty, mice homozygous null for REA in the mammary gland (REAf/f PRcre/+) showed severely impaired mammary ductal elongation and morphogenesis, whereas mice heterozygous for REA (REAf/+ PRcre/+) displayed accelerated mammary ductal elongation, increased numbers of terminal end buds, and up-regulation of amphiregulin, the major paracrine mediator of estrogen-induced ductal morphogenesis. During pregnancy and lactation, mice with homozygous REA gene deletion in mammary epithelium (REAf/f whey acidic protein-Cre) showed a loss of lobuloalveolar structures and increased apoptosis of mammary alveolar epithelium, leading to impaired milk production and significant reduction in growth of their offspring, whereas body weights of the offspring nursed by females heterozygous for REA were slightly greater than those of control mice. Our findings reveal that REA is essential for mammary gland development and has a gene dosage-dependent role in the regulation of stage-specific physiological functions of the mammary gland.


Subject(s)
Lactation/genetics , Mammary Glands, Animal/growth & development , Morphogenesis/physiology , Repressor Proteins/genetics , Animals , Apoptosis/genetics , Epithelial Cells/metabolism , Female , Gene Dosage , Lactation/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Pregnancy , Prohibitins , Repressor Proteins/metabolism
8.
Biol Reprod ; 85(2): 386-96, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21508348

ABSTRACT

The success of postnatal uterine morphogenesis dictates, in part, the embryotrophic potential and functional capacity of the adult uterus. The definitive role of Wnt7a in postnatal uterine development and adult function requires a conditional knockout, because global deletion disrupts müllerian duct patterning, specification, and cell fate in the fetus. The Wnt7a-null uterus appears to be posteriorized because of developmental defects in the embryo, as evidenced by the stratified luminal epithelium that is normally found in the vagina and the presence of short and uncoiled oviducts. To understand the biological role of WNT7A after birth and allow tissue-selective deletion of Wnt7a, we generated loxP-flanked exon 2 mice and conditionally deleted Wnt7a after birth in the uterus by crossing them with Pgr(Cre) mice. Morphological examination revealed no obvious differences in the vagina, cervix, oviduct, or ovary. The uteri of Wnt7a mutant mice contained no endometrial glands, whereas all other uterine cell types appeared to be normal. Postnatal differentiation of endometrial glands was observed in control mice, but not in mutant mice, between Postnatal Days 3 and 12. Expression of morphoregulatory genes, particularly Foxa2, Hoxa10, Hoxa11, Msx1, and Wnt16, was disrupted in the Wnt7a mutant uteri. Conditional Wnt7a mutant mice were not fertile. Although embryos were present in uteri of mutant mice on Day 3.5 of pregnancy, blastocyst implantation was not observed on Day 5.5. Furthermore, expression of several genes (Foxa2, Lif, Msx1, and Wnt16) was reduced or absent in adult Wnt7a-deleted uteri on Day 3.5 postmating. These results indicate that WNT7A plays a critical role in postnatal uterine gland morphogenesis and function, which are important for blastocyst implantation and fertility in the adult uterus.


Subject(s)
Fertility/physiology , Gene Deletion , Uterus/growth & development , Wnt Proteins/metabolism , Animals , Cell Proliferation , Embryo Implantation/physiology , Female , Fertility/genetics , Gene Expression Regulation/physiology , Mice , Uterus/cytology , Uterus/metabolism , Wnt Proteins/genetics
9.
J Clin Endocrinol Metab ; 96(6): 1737-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411543

ABSTRACT

CONTEXT: Polycystic ovary syndrome (PCOS), the most common endocrinopathy of reproductive-aged women, is characterized by ovulatory dysfunction and hyperandrogenism. OBJECTIVE: The aim was to compare gene expression between endometrial samples of normal fertile controls and women with PCOS. DESIGN AND SETTING: We conducted a case control study at university teaching hospitals. PATIENTS: Normal fertile controls and women with PCOS participated in the study. INTERVENTIONS: Endometrial samples were obtained from normal fertile controls and from women with PCOS, either induced to ovulate with clomiphene citrate or from a modeled secretory phase using daily administration of progesterone. MAIN OUTCOME MEASURE: Total RNA was isolated from samples and processed for array hybridization with Affymetrix HG U133 Plus 2 arrays. Data were analyzed using GeneSpring GX11 and Ingenuity Pathways Analysis. Selected gene expression differences were validated using RT-PCR and/or immunohistochemistry in separately obtained PCOS and normal endometrium. RESULTS: ANOVA analysis revealed 5160 significantly different genes among the three conditions. Of these, 466 were differentially regulated between fertile controls and PCOS. Progesterone-regulated genes, including mitogen-inducible gene 6 (MIG6), leukemia inhibitory factor (LIF), GRB2-associated binding protein 1 (GAB1), S100P, and claudin-4 were significantly lower in PCOS endometrium; whereas cell proliferation genes, such as Anillin and cyclin B1, were up-regulated. CONCLUSIONS: Differences in gene expression provide evidence of progesterone resistance in midsecretory PCOS endometrium, independent of clomiphene citrate and corresponding to the observed phenotypes of hyperplasia, cancer, and poor reproductive outcomes in this group of women.


Subject(s)
Clomiphene/therapeutic use , Endometrium/metabolism , Polycystic Ovary Syndrome/metabolism , Progesterone/therapeutic use , Analysis of Variance , Case-Control Studies , Clomiphene/pharmacology , Endometrium/drug effects , Female , Gene Expression/drug effects , Gene Expression/physiology , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Principal Component Analysis , Progesterone/pharmacology
10.
Cell Metab ; 13(1): 35-43, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21195347

ABSTRACT

All organisms have devised strategies to counteract energy depletion and promote fitness for survival. We show here that cellular energy depletion puts into play a surprising strategy that leads to absorption of exogenous fuel for energy repletion. The energy-depletion-sensing kinase AMPK binds, phosphorylates, and activates the transcriptional coactivator SRC-2, which in a liver-specific manner promotes absorption of dietary fat from the gut. Hepatocyte-specific deletion of SRC-2 results in intestinal fat malabsorption and attenuated entry of fat into the blood stream. This defect can be attributed to AMPK- and SRC-2-mediated transcriptional regulation of hepatic bile acid (BA) secretion into the gut, as it can be completely rescued by replenishing intestinal BA or by genetically restoring the levels of hepatic bile salt export pump (BSEP). Our results position the hepatic AMPK-SRC-2 axis as an energy rheostat, which upon cellular energy depletion resets whole-body energy by promoting absorption of dietary fuel.


Subject(s)
AMP-Activated Protein Kinases/metabolism , ATP-Binding Cassette Transporters/metabolism , Dietary Fats/metabolism , Nuclear Receptor Coactivator 2/deficiency , Nuclear Receptor Coactivator 2/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Ablation Techniques , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Energy Metabolism , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Intestinal Absorption , Liver/cytology , Liver/enzymology , Liver/metabolism , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Male , Mice , Mice, Knockout , Nuclear Receptor Coactivator 2/genetics , Phosphorylation , Promoter Regions, Genetic , RNA-Binding Proteins/metabolism , Transcriptional Activation
11.
Cell Metab ; 12(6): 606-18, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21109193

ABSTRACT

Gluconeogenesis makes a major contribution to hepatic glucose production, a process critical for survival in mammals. In this study, we identify the p160 family member, SRC-1, as a key coordinator of the hepatic gluconeogenic program in vivo. SRC-1-null mice displayed hypoglycemia secondary to a deficit in hepatic glucose production. Selective re-expression of SRC-1 in the liver restored blood glucose levels to a normal range. SRC-1 was found induced upon fasting to coordinate in a cell-autonomous manner, the gene expression of rate-limiting enzymes of the gluconeogenic pathway. At the molecular level, the main role of SRC-1 was to modulate the expression and the activity of C/EBPα through a feed-forward loop in which SRC-1 used C/EBPα to transactivate pyruvate carboxylase, a crucial gene for initiation of the gluconeogenic program. We propose that SRC-1 acts as a critical mediator of glucose homeostasis in the liver by adjusting the transcriptional activity of key genes involved in the hepatic glucose production machinery.


Subject(s)
Gene Expression Regulation/physiology , Gluconeogenesis/physiology , Glucose/biosynthesis , Hypoglycemia/metabolism , Liver/metabolism , Nuclear Receptor Coactivator 1/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Gene Expression Profiling , Immunoprecipitation , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction
12.
Science ; 322(5906): 1395-9, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-19039140

ABSTRACT

Hepatic glucose production is critical for basal brain function and survival when dietary glucose is unavailable. Glucose-6-phosphatase (G6Pase) is an essential, rate-limiting enzyme that serves as a terminal gatekeeper for hepatic glucose release into the plasma. Mutations in G6Pase result in Von Gierke's disease (glycogen storage disease-1a), a potentially fatal genetic disorder. We have identified the transcriptional coactivator SRC-2 as a regulator of fasting hepatic glucose release, a function that SRC-2 performs by controlling the expression of hepatic G6Pase. SRC-2 modulates G6Pase expression directly by acting as a coactivator with the orphan nuclear receptor RORalpha. In addition, SRC-2 ablation, in both a whole-body and liver-specific manner, resulted in a Von Gierke's disease phenotype in mice. Our results position SRC-2 as a critical regulator of mammalian glucose production.


Subject(s)
Glucose-6-Phosphatase/genetics , Glucose/metabolism , Glycogen Storage Disease Type I/genetics , Liver/metabolism , Nuclear Receptor Coactivator 2/metabolism , Animals , Cells, Cultured , Fasting , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/metabolism , Hepatocytes/metabolism , Kidney/metabolism , Liver Glycogen/metabolism , Male , Mice , Mice, Knockout , Nuclear Receptor Coactivator 2/genetics , RNA Interference , Receptors, Retinoic Acid/metabolism , Response Elements , Retinoic Acid Receptor alpha , Transcription, Genetic , Triglycerides/metabolism
13.
Proc Natl Acad Sci U S A ; 102(38): 13490-5, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16150722

ABSTRACT

We previously reported that an Nkx2-5-GFP bacterial artificial chromosome in transgenic mice recapitulated the endogenous gene activity in the heart. Here, we identified three additional previously uncharacterized distal enhancer modules of Nkx2-5: UH6, which directed transgene expression in the right ventricle, interventricular septum, and atrial ventricular canal; UH5, which directed expression in both atria; and UH4, which directed transgene expression in tongue muscle. Nkx2-5 enhancers drive cardiogenic gene activity from the earliest progenitors to the late-stage embryonic heart, reside within its 27 kb of 5' flanking sequences, organized in a tandem array. Nkx2-5 enhancers involved with stomach-, tongue-, and chamber-restricted expression displayed lacZ transgene activity and chromatin histone acetylation patterns consistent with tissue-specific expression. An examination of Nkx2-5 gene activity in murine embryonic stem cells converted to beating embryoid bodies showed that only the proximal active region 2 and GATA-Smad enhancers were chromatin-remodeled. Chromatin remodeling of active region 2 and GATA-Smad enhancers were blunted by noggin coexpression, which indicated dependence on bone morphogenetic protein signaling for their chromatin activation during activation of Nkx2-5 expression.


Subject(s)
Carrier Proteins/metabolism , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Chromosomes, Artificial, Bacterial/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Heart Atria/embryology , Heart Ventricles/embryology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Organ Specificity/genetics , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism , Tongue/embryology , Transcription Factors/genetics , Transgenes/physiology
14.
Exp Lung Res ; 31(1): 37-55, 2005.
Article in English | MEDLINE | ID: mdl-15765918

ABSTRACT

Constitutive expression of human achaete-scute homolog-1 (hASH-1) in combination with simian virus large Tantigen under the Clara cell 10-kDa secretory protein (CC10) promoter results in adenocarcinomas with focal neuroendocrine (NE) differentiation. Mice carrying conditional alleles for both Rb-1 and p53 in lung epithelial cells develop aggressive lung tumors with similarities to human small cell lung cancers, including high level expression of ASH-1, NE markers, and extra-pulmonary metastases. Tumors in both models originate from bronchiolar epithelium, reveal a range of premalignant changes, express thyroid transcription factor-1 (TTF-1), a marker of peripheral airway cell lineage, and display varying degrees of bidirectional epithelial/NE differentiation.


Subject(s)
Carcinoma, Neuroendocrine/secondary , Carcinoma, Non-Small-Cell Lung/secondary , Disease Models, Animal , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Transcription Factors/genetics , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Basic Helix-Loop-Helix Transcription Factors , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uteroglobin/genetics , Uteroglobin/metabolism
15.
Fertil Steril ; 82(3): 673-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15374713

ABSTRACT

OBJECTIVE: To determine the role of P and its nuclear receptor PR in the growth of ectopic uterine tissue of mice with or without a disrupted PR gene. DESIGN: Animal study. SETTING: Academic medical center. ANIMAL(S): Female wild-type (WT) and transgenic knockout mice for P receptor (PRKO). INTERVENTION(S): Endometriosis was induced in the following groups of ovariectomized adult mice: [1] untreated WT, [2] estradiol (E(2))-treated WT, [3] P-treated WT, [4] E(2) + P-treated WT, [5] untreated PRKO, [6] E(2)-treated PRKO, and [7] E(2) + P-treated PRKO (n = 5 in each group). MAIN OUTCOME MEASURE(S): The size of ectopic uterine tissue in WT and PRKO mice were compared between the groups subjected to treatments with P or E(2). Tissue proliferating cell nuclear antigen (PCNA) levels were compared among these groups. RESULTS: Treatment with P only significantly decreased the size of WT ectopic uterine tissue. The untreated PRKO ectopic uterine tissue was significantly larger than WT tissue. Estradiol increased the size of ectopic uterine tissues, and this E(2)-dependent growth could be suppressed by P in WT tissues but not in PRKO tissues. Finally, the hormone-dependent changes in ectopic uterine tissue size were accompanied by comparable alterations in PCNA levels. CONCLUSION(S): Intact PR in ectopic uterine tissue is essential to abolish E(2)-dependent or -independent proliferation. We also suggest that ectopic uterine tissue is associated with significantly increased resistance to P action and increased predisposition to E(2)-dependent proliferation in the absence of PR. Overall, these findings support the hypothesis that P resistance in human endometriosis may be due to the absence of sufficient levels of functional PR in this tissue.


Subject(s)
Cell Division/physiology , Endometriosis/pathology , Estradiol/pharmacology , Receptors, Progesterone/physiology , Animals , Cell Division/drug effects , Disease Models, Animal , Endometriosis/genetics , Female , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics
16.
Differentiation ; 70(9-10): 624-32, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12492503

ABSTRACT

The minimal rat probasin (PB) promoter was used to target expression of human fibroblast growth factor-7 (FGF-7)/keratinocyte growth factor (KGF) directly to prostatic epithelium of transgenic mice, converting FGF-7 from a paracrine to an autocrine factor. Four independent lines were established that expressed the transgene (PKS) in the prostate. Upon histologic analysis, the prostatic epithelium of PKS mice was found to be hyperplastic. Many of the prostatic ducts were filled with secretory epithelial cells tightly associated with a highly enfolded basement membrane. Distortions of the ductal smooth muscle layer were also observed. Prostates from year-old PKS mice had significantly more abnormal ducts than their wild-type nontransgenic littermates. The minimal rat PB promoter was also used to target a truncated FGFR2iiib receptor to prostatic epithelium to functionally abrogate endogenous FGF-7 signaling. Three lines were established that expressed the transgene (KDNR) in the prostate. Upon dissection it was noted that all four lobes of the prostates of KDNR mice were present but smaller in size. Histologic analysis indicated that the epithelium in many of the prostatic ducts was disorganized and contained numerous rounded cytokeratin-positive cells that were not tightly associated with the basement membrane. The stroma was disorganized and did not form a tight layer of smooth muscle around the epithelial ducts. Surprisingly, abrogation of FGF signaling in KDNR mice correlated with the emergence of a neuroendocrine-like phenotype that was not observed as a consequence of enforced FGF-7 expression in the PKS mice.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors/genetics , Prostate/pathology , Prostatic Hyperplasia/genetics , Receptors, Fibroblast Growth Factor/genetics , Androgen-Binding Protein/genetics , Animals , Autocrine Communication/genetics , Cell Differentiation/genetics , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/metabolism , Humans , Male , Mice , Mice, Transgenic , Paracrine Communication/genetics , Promoter Regions, Genetic , Prostate/physiology , Prostatic Hyperplasia/etiology , Prostatic Hyperplasia/pathology , Rats , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...