Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
F S Sci ; 5(1): 80-91, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38043603

ABSTRACT

OBJECTIVES: To assess the effect of simvastatin on uterine leiomyoma growth and extracellular matrix (ECM) deposition. DESIGN: Laboratory analysis of human leiomyoma cell culture, xenograft in a mouse model, and patient tissue from a clinical trial. SETTING: Academic research center. PATIENT(S): Tissue culture from human leiomyoma tissue and surgical leiomyoma tissue sections from a placebo-controlled randomized clinical trial. INTERVENTION(S): Simvastatin treatment. MAIN OUTCOME MEASURE(S): Serum concentrations, xenograft volumes, and protein expression. RESULTS: Mice xenografted with 3-dimensional human leiomyoma cultures were divided as follows: 7 untreated controls; 12 treated with activated simvastatin at 10 mg/kg body weight; and 15 at 20 mg/kg body weight. Simvastatin was detected in the serum of mice injected at the highest dose. Xenograft volumes were significantly smaller (mean 53% smaller at the highest concentration). There was dissolution of compact ECM, decreased ECM formation, and lower collagen protein expression in xenografts. Membrane type 1 matrix metalloproteinase was increased in vitro and in vivo. Matrix metalloproteinase 2 and low-density lipoprotein receptor-related protein 1 were increased in vitro. CONCLUSIONS: Simvastatin exhibited antitumoral activity with ECM degradation and decreased leiomyoma tumor volume in vivo. Activation of the matrix metalloproteinase 2, membrane type 1 matrix metalloproteinase, and low-density lipoprotein receptor-related protein 1 pathway may explain these findings.


Subject(s)
Leiomyoma , Uterine Neoplasms , Female , Humans , Mice , Animals , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Simvastatin/pharmacology , Simvastatin/metabolism , Simvastatin/therapeutic use , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Leiomyoma/drug therapy , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Body Weight , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/therapeutic use
2.
F S Sci ; 4(1): 74-89, 2023 02.
Article in English | MEDLINE | ID: mdl-36273722

ABSTRACT

OBJECTIVE: To determine whether a curcumin-supplemented diet would prevent and/or treat uterine leiomyoma growth in our mouse xenograft model. DESIGN: Animal study. SETTING: Laboratory study. PATIENT(S): N/A. INTERVENTION(S): Curcumin-supplemented diet. MAIN OUTCOME MEASURE(S): Dietary intake, blood concentrations, tumor size, extracellular matrix protein concentrations, apoptosis markers. RESULT(S): We found that curcumin was well tolerated as a dietary supplement, free curcumin and its metabolites were detected in the serum, and exposure resulted in approximately 60% less leiomyoma xenograft growth as well as dissolution of the peripheral extracellular matrix architecture of the xenografts. The production of matrix proteins, including collagens, decreased, whereas the number of apoptotic cells in the xenografts increased. Additionally, when xenografts were placed in a uterine intramural location, we found a significantly increased apoptotic response to curcumin in the diet. CONCLUSION(S): Mice on a diet supplemented with curcumin could achieve serum concentrations sufficient to regulate human leiomyoma xenograft growth, and curcumin could play both preventive and curative roles in the treatment of uterine leiomyoma as an oral nutritional supplement.


Subject(s)
Curcumin , Leiomyoma , Uterine Neoplasms , Female , Humans , Animals , Mice , Curcumin/pharmacology , Curcumin/therapeutic use , Curcumin/metabolism , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Heterografts , Solubility , Leiomyoma/drug therapy , Extracellular Matrix/metabolism , Extracellular Matrix/pathology
3.
Reprod Sci ; 30(5): 1528-1539, 2023 05.
Article in English | MEDLINE | ID: mdl-36401072

ABSTRACT

Granulosa cells (GCs) must respond appropriately to follicle-stimulating hormone (FSH) for proper follicle maturation. FSH activates protein kinase A (PKA) leading to phosphorylation of the cyclic AMP response element binding protein-1 (CREB1). We identified a unique A-kinase anchoring protein (AKAP13) containing a Rho guanine nucleotide exchange factor (RhoGEF) region that was induced in GCs during folliculogenesis. AKAPs are known to coordinate signaling cascades, and we sought to evaluate the role of AKAP13 in GCs in response to FSH. Aromatase reporter activity was increased in COV434 human GCs overexpressing AKAP13. Addition of FSH, or the PKA activator forskolin, significantly enhanced this activity by 1.5- to 2.5-fold, respectively (p < 0.001). Treatment with the PKA inhibitor H89 significantly reduced AKAP13-dependent activation of an aromatase reporter (p = 0.0067). AKAP13 physically interacted with CREB1 in co-immunoprecipitation experiments and increased the phosphorylation of CREB1. CREB1 phosphorylation increased after FSH treatment in a time-specific manner, and this effect was reduced by siRNA directed against AKAP13 (p = 0.05). CREB1 activation increased by 18.5-fold with co-expression of AKAP13 in the presence of FSH (p < 0.001). Aromatase reporter activity was reduced by inhibitors of the RhoGEF region, C3 transferase and A13, and greatly enhanced by the RhoGEF activator, A02. In primary murine and COV43 GCs, siRNA knockdown of Akap13/AKAP13 decreased aromatase and luteinizing hormone receptor transcripts in cells treated with FSH, compared with controls. Collectively, these findings suggest that AKAP13 may function as a scaffolding protein in FSH signal transduction via an interaction with CREB, resulting in phosphorylation of CREB.


Subject(s)
A Kinase Anchor Proteins , Follicle Stimulating Hormone , Female , Humans , Mice , Animals , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , A Kinase Anchor Proteins/metabolism , A Kinase Anchor Proteins/pharmacology , Aromatase/metabolism , Granulosa Cells/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone, Human/pharmacology , Rho Guanine Nucleotide Exchange Factors/metabolism , Cells, Cultured , Proto-Oncogene Proteins/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism
4.
F S Sci ; 3(2): 118-129, 2022 05.
Article in English | MEDLINE | ID: mdl-35560009

ABSTRACT

OBJECTIVE: To determine whether the mechanotransduction and pharmacomanipulation of A-kinase anchoring protein 13 (AKAP13) altered Hippo signaling pathway transcription and growth factors in granulosa cells. Primary ovarian insufficiency is the depletion or dysfunction of primordial ovarian follicles. In vitro activation of ovarian tissue in patients with primary ovarian insufficiency alters the Hippo and phosphatase and tensin homolog/phosphatidylinositol 3-kinase/protein kinase B/forkhead box O3 pathways. A-kinase anchoring protein 13 is found in granulosa cells and may regulate the Hippo pathway via F-actin polymerization resulting in altered nuclear yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif coactivators and Tea domain family (TEAD) transcription factors. DESIGN: Laboratory studies. SETTING: Translational science laboratory. PATIENT(S): None. INTERVENTION(S): COV434 cells, derived from a primary human granulosa tumor cell line, were studied under different cell density and well stiffness conditions. Cells were transfected with a TEAD-luciferase (TEAD-luc) reporter as well as expression constructs for AKAP13 or AKAP13 mutants and then treated with AKAP13 activators, inhibitors, and follicle-stimulating hormone. MAIN OUTCOME MEASURE(S): TEAD gene activation or inhibition was measured by TEAD-luciferase assays. The messenger ribonucleic acid levels of Hippo pathway signaling molecules, including connective tissue growth factor (CTGF), baculoviral inhibitors of apoptosis repeat-containing 5, Ankyrin repeat domain-containing protein 1, YAP1, and TEAD1, were measured by quantitative real-time polymerase chain reaction. Protein expressions for AKAP13, CTGF, YAP1, and TEAD1 were measured using Western blot. RESULT(S): Increased TEAD-luciferase activity and expression of markers for cellular growth were associated with decreased cell density, increased well stiffness, and AKAP13 activator (A02) treatment. Additionally, decreased TEAD-luc activity and expression of markers for cellular growth were associated with AKAP13 inhibitor (A13) treatment, including a reduced expression of the BIRC5 and ANKRD1 (YAP-responsive genes) transcript levels and CTGF protein levels. There were no changes in TEAD-luc with follicle-stimulating hormone treatment, supporting Hippo pathway involvement in the gonadotropin-independent portion of folliculogenesis. CONCLUSION(S): These findings suggest that AKAP13 mediates Hippo-regulated changes in granulosa cell growth via mechanotransduction and pharmacomanipulation. The AKAP13 regulation of the Hippo pathway may represent a potential target for regulation of follicle activation.


Subject(s)
Primary Ovarian Insufficiency , Protein Serine-Threonine Kinases , A Kinase Anchor Proteins/metabolism , Female , Follicle Stimulating Hormone/metabolism , Hippo Signaling Pathway , Humans , Mechanotransduction, Cellular , Ovarian Follicle , Primary Ovarian Insufficiency/metabolism , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics
5.
Sci Rep ; 11(1): 9371, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931688

ABSTRACT

Pathogenic mutations in fumarate hydratase (FH) drive hereditary leiomyomatosis and renal cell cancer (HLRCC) and increase the risk of developing uterine leiomyomas (ULMs). An integrated proteogenomic analysis of ULMs from HLRCC (n = 16; FH-mutation confirmed) and non-syndromic (NS) patients (n = 12) identified a significantly higher protein:transcript correlation in HLRCC (R = 0.35) vs. NS ULMs (R = 0.242, MWU p = 0.0015). Co-altered proteins and transcripts (228) included antioxidant response element (ARE) target genes, such as thioredoxin reductase 1 (TXNRD1), and correlated with activation of NRF2-mediated oxidative stress response signaling in HLRCC ULMs. We confirm 185 transcripts previously described as altered between HLRCC and NS ULMs, 51 co-altered at the protein level and several elevated in HLRCC ULMs are involved in regulating cellular metabolism and glycolysis signaling. Furthermore, 367 S-(2-succino)cysteine peptides were identified in HLRCC ULMs, of which sixty were significantly elevated in HLRCC vs. NS ULMs (LogFC = 1.86, MWU p < 0.0001). These results confirm and define novel proteogenomic alterations in uterine leiomyoma tissues collected from HLRCC patients and underscore conserved molecular alterations correlating with inactivation of the FH tumor suppressor gene.


Subject(s)
Biomarkers, Tumor/analysis , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Leiomyomatosis/pathology , Mutation , Neoplastic Syndromes, Hereditary/pathology , Proteogenomics/methods , Proteome/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Case-Control Studies , Female , Humans , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Proteome/analysis , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism
6.
F S Sci ; 2(4): 396-406, 2021 11.
Article in English | MEDLINE | ID: mdl-35559862

ABSTRACT

OBJECTIVE: To assess and characterize the role of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in the development of uterine leiomyoma. DESIGN: Laboratory study. SETTING: Academic research center. PATIENT(S): Not applicable. INTERVENTION(S): Laboratory investigation. In vitro assessment of human leiomyoma and myometrial tissue specimens as well as immortalized leiomyoma and myometrial cell lines. MAIN OUTCOME MEASURE(S): Western blotting and immunohistochemical analyses were performed to assess differences in CEACAM1 content between leiomyoma and myometrial samples. Small interfering RNA silencing experiments and transient transfection experiments were performed to characterize the regulatory role of CEACAM1 on downstream signaling cascades. RESULT(S): Analysis of RNA sequencing data revealed decreased CEACAM1 expression in human uterine leiomyoma specimens compared with that in myometrial samples. This translated to a significant down-regulation in CEACAM1 protein content in human leiomyoma compared with patient-matched myometrial tissue samples (0.236 ± 0.05-fold). A similar decrease in CEACAM1 protein content was observed in matched immortalized leiomyoma cell (ILC) and immortalized myometrial cell lines (0.21 ± 0.07). Immunohistochemical analysis revealed decreased staining intensity in leiomyoma surgical specimens compared with the matched myometrium of placebo patients. Lower CEACAM1 levels in leiomyoma were associated with increased activation of both the mitogen-activated protein kinase (MAPK) and the phosphoinositide 3-kinase/protein kinase B pathways compared with that in myometrial cells. This is significant because activation of these pathways plays an important role in leiomyoma growth. Treatment of myometrial cells with CEACAM1 small interfering RNA resulted in a significant down-regulation of CEACAM1 at the protein level (0.272 ± 0.06-fold) and was associated with increased activation of the MAPK (1.62 ± 0.21-fold) and phosphoinositide 3-kinase/protein kinase B (1.79 ± 0.35-fold) pathways, as well as increased collagen production (2.1 ± 0.49-fold). Rescue of CEACAM1 expression in leiomyoma cells by transient transfection restored regulatory control and resulted in lower activation of the MAPK pathway (0.58 ± 0.37-fold). CONCLUSION(S): CEACAM1 is an important protein involved in regulating many signal transduction pathways. Decreased CEACAM1 expression in leiomyoma allows permissive uncontrolled overactivation and up-regulation of downstream pathways that may contribute to leiomyoma growth.


Subject(s)
Leiomyoma , Uterine Neoplasms , Antigens, CD , Carcinoembryonic Antigen/therapeutic use , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Female , Humans , Leiomyoma/genetics , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/therapeutic use , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/therapeutic use , Uterine Neoplasms/genetics
7.
F S Sci ; 2(3): 303-314, 2021 08.
Article in English | MEDLINE | ID: mdl-35560280

ABSTRACT

OBJECTIVE: To determine if A-kinase anchoring protein 13 (AKAP13) interacts with the vitamin D receptor (VDR) to alter vitamin D-dependent signaling in fibroid cells. Uterine leiomyomas (fibroids) are characterized by a fibrotic extracellular matrix and are associated with vitamin D deficiency. Treatment with vitamin D (1,25-dihydroxyvitamin D3) reduces fibroid growth and extracellular matrix gene expression. A-kinase anchoring protein 13 is overexpressed in fibroids and interacts with nuclear hormone receptors, but it is not known whether AKAP13 may interact with the VDR to affect vitamin D signaling in fibroids. DESIGN: Laboratory studies. SETTING: Translational science laboratory. INTERVENTION(S): Human immortalized fibroid or myometrial cells were treated with 1,25-hydroxyvitamin D3 (1,25(OH)2D3) and transfected using expression constructs for AKAP13 or AKAP13 mutants, RhoQL, C3 transferase, or small interfering ribonucleic acids (RNAs). MAIN OUTCOME MEASURE(S): Messenger ribonucleic acid (mRNA) levels of AKAP13, fibromodulin, and versican as measured by quantitative real-time polymerase chain reaction. Glutathione S-transferase-binding assays. Vitamin D-dependent gene activation as measured by luciferase assays. RESULT(S): 1,25(OH)2D3 resulted in a significant reduction in mRNA levels encoding AKAP13, versican, and fibromodulin. Small interfering RNA silencing of AKAP13 decreased both fibromodulin and versican mRNA levels. Glutathione S-transferase-binding assays revealed that AKAP13 bound to the VDR through its nuclear receptor interacting region. Cotransfection of AKAP13 and VDR significantly reduced vitamin D-dependent gene activation. RhoA pathway inhibition partially relieved repression of vitamin D-dependent gene activation by AKAP13. CONCLUSION(S): These data suggest that AKAP13 inhibited the vitamin D receptor activation by a mechanism that required, at least in part, RhoA activation.


Subject(s)
Leiomyoma , Receptors, Calcitriol , A Kinase Anchor Proteins/genetics , Female , Fibromodulin/genetics , Glutathione Transferase/genetics , Humans , Leiomyoma/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Transcriptional Activation , Versicans/genetics , Vitamin D/pharmacology , Vitamins
8.
J Clin Endocrinol Metab ; 104(3): 970-980, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30239831

ABSTRACT

Context: Uterine leiomyomata (fibroids) are prevalent sex hormone‒dependent tumors with an altered response to mechanical stress. Ulipristal acetate, a selective progesterone receptor (PR) modulator, significantly reduces fibroid size in patients. However, PR signaling in fibroids and its relationship to mechanical signaling are incompletely understood. Objective: Our prior studies revealed that A-kinase anchoring protein 13 (AKAP13) was overexpressed in fibroids and contributed to altered mechanotransduction in fibroids. Because AKAP13 augmented nuclear receptor signaling in other tissues, we sought to determine whether AKAP13 might influence PR signaling in fibroids. Methods and Results: Fibroid samples from patients treated with ulipristal acetate or placebo were examined for AKAP13 expression by using immunohistochemistry. In immortalized uterine fibroid cell lines and COS-7 cells, we observed that AKAP13 increased ligand-dependent PR activation of luciferase reporters and endogenous progesterone-responsive genes for PR-B but not PR-A. Inhibition of ERK reduced activation of PR-dependent signaling by AKAP13, but inhibition of p38 MAPK had no effect. In addition, glutathione S-transferase‒binding assays revealed that AKAP13 was bound to PR-B through its carboxyl terminus. Conclusion: These data suggest an intersection of mechanical signaling and PR signaling involving AKAP13 through ERK. Further elucidation of the integration of mechanical and hormonal signaling pathways in fibroids may provide insight into fibroid development and suggest new therapeutic strategies for treatment.


Subject(s)
A Kinase Anchor Proteins/metabolism , Leiomyoma/pathology , Minor Histocompatibility Antigens/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Progesterone/metabolism , Uterine Neoplasms/pathology , A Kinase Anchor Proteins/genetics , Adult , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Female , Gene Knockdown Techniques , Humans , Leiomyoma/drug therapy , MAP Kinase Signaling System/drug effects , Mechanotransduction, Cellular/drug effects , Middle Aged , Minor Histocompatibility Antigens/genetics , Norpregnadienes/pharmacology , Norpregnadienes/therapeutic use , Progesterone/metabolism , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/metabolism , Receptors, Progesterone/antagonists & inhibitors , Uterine Neoplasms/drug therapy , Uterus/drug effects , Uterus/pathology
9.
Trends Endocrinol Metab ; 28(9): 656-668, 2017 09.
Article in English | MEDLINE | ID: mdl-28651856

ABSTRACT

Progesterone is essential for pregnancy maintenance and menstrual cycle regulation. Hormone action has been primarily ascribed to the well-characterized classical signaling pathway involving ligand binding, activation of nuclear progesterone receptors (PRs), and subsequent activation of genes containing progesterone response elements (PREs). Recent studies have revealed progesterone actions via non-classical signaling pathways, often mediated by non-genomic signaling. Progesterone signaling, in conjunction with growth factor signaling, impacts on the function of growth factors and regulates important physiological actions such as cell growth and remodeling, as well as apoptosis. This review focuses on non-classical progesterone signaling pathways, both including and excluding PR, and highlights how research in this area will provide a better understanding of progesterone actions and may inform novel therapeutic strategies.


Subject(s)
Progesterone/metabolism , Progesterone/pharmacology , Animals , Humans , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Receptors, Progesterone/metabolism , Receptors, Progesterone/physiology , Signal Transduction/drug effects
10.
Fertil Steril ; 104(6): 1344-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26602982

ABSTRACT

The process of reproduction inherently poses unique microbial challenges because it requires the transfer of gametes from one individual to the other, meanwhile preserving the integrity of the gametes and individuals from harmful microbes during the process. Advances in molecular biology techniques have expanded our understanding of the natural organisms living on and in our bodies, including those inhabiting the reproductive tract. Over the past two decades accumulating evidence has shown that the human microbiome is tightly related to health and disease states involving the different body systems, including the reproductive system. Here we introduce the science involved in the study of the human microbiome. We examine common methods currently used to characterize the human microbiome as an inseparable part of the reproductive system. Finally, we consider a few limitations, clinical implications, and the critical need for additional research in the field of human fertility.


Subject(s)
Bacteria/genetics , DNA, Bacterial/genetics , Genitalia/microbiology , Microbiota , Molecular Diagnostic Techniques , Reproduction , Ribotyping , Bacteria/classification , Bacteria/growth & development , Bacteria/isolation & purification , Computational Biology , DNA, Bacterial/isolation & purification , Dysbiosis , Female , Genitalia/physiopathology , Host-Pathogen Interactions , Humans , Infertility/microbiology , Infertility/physiopathology , Male
11.
J Bone Miner Res ; 30(10): 1887-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25892096

ABSTRACT

Mechanical stimulation is crucial to bone growth and triggers osteogenic differentiation through a process involving Rho and protein kinase A. We previously cloned a gene (AKAP13, aka BRX) encoding a protein kinase A-anchoring protein in the N-terminus, a guanine nucleotide-exchange factor for RhoA in the mid-section, coupled to a carboxyl region that binds to estrogen and glucocorticoid nuclear receptors. Because of the critical role of Rho, estrogen, and glucocorticoids in bone remodeling, we examined the multifunctional role of Akap13. Akap13 was expressed in bone, and mice haploinsufficient for Akap13 (Akap13(+/-)) displayed reduced bone mineral density, reduced bone volume/total volume, and trabecular number, and increased trabecular spacing; resembling the changes observed in osteoporotic bone. Consistent with the osteoporotic phenotype, Colony forming unit-fibroblast numbers were diminished in Akap13(+/-) mice, as were osteoblast numbers and extracellular matrix production when compared to control littermates. Transcripts of Runx2, an essential transcription factor for the osteogenic lineage, and alkaline phosphatase (Alp), an indicator of osteogenic commitment, were both reduced in femora of Akap13(+/-) mice. Knockdown of Akap13 reduced levels of Runx2 and Alp transcripts in immortalized bone marrow stem cells. These findings suggest that Akap13 haploinsufficient mice have a deficiency in early osteogenesis with a corresponding reduction in osteoblast number, but no impairment of mature osteoblast activity.


Subject(s)
A Kinase Anchor Proteins/deficiency , Bone Density , Guanine Nucleotide Exchange Factors/deficiency , Osteoporosis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Haploinsufficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
12.
J Steroid Biochem Mol Biol ; 122(4): 279-86, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20493256

ABSTRACT

Uterine leiomyomas are highly prevalent and often symptomatic, but current medical therapies are limited. A novel, potent, selective, orally active therapy is needed. The goal of these studies was to determine the progesterone receptor (PR) specificity and activation, endometrial response, and impact on leiomyoma cell proliferation and extracellular matrix (ECM) production of the novel non-steroidal selective progesterone receptor modulators (SPRMs) CP8863 and CP8947. In vitro progestational activity was assessed by alkaline phosphatase assay and ER-α expression. In vivo progestational activity was assayed by the McPhail assay. Proliferation and gene expression studies were performed in immortalized human leiomyoma and myometrial cells. Both CP8863 and CP8947 were highly selective for progesterone receptor (PR) but not for ER-α, AR, and GR. Both compounds induced alkaline phosphatase comparably to progesterone, while CP8947 induced ER-α in leiomyoma cells but not myometrial cells. CP8947 was progestational in rabbit endometrium. Nanomolar CP8947 treatment inhibited human leiomyoma but not myometrial cell proliferation. Extracellular matrix components were decreased in leiomyoma cells, including COL1A1 and COL7A1 at nanomolar concentrations. CP8947 was a potent novel non-steroidal SPRM that was selective for PR, demonstrated progestational activity in endometrium, inhibited leiomyoma cell proliferation and decreased ECM component production, without disrupting myometrial cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Indoles/pharmacology , Leiomyoma/drug therapy , Receptors, Progesterone/metabolism , Uterine Neoplasms/drug therapy , Animals , Cell Line, Tumor , Endometrium/drug effects , Female , Humans , Myometrium/drug effects , Rabbits
13.
J Biol Chem ; 285(16): 12344-54, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20139090

ABSTRACT

A fundamental biologic principle is that diverse biologic signals are channeled through shared signaling cascades to regulate development. Large scaffold proteins that bind multiple proteins are capable of coordinating shared signaling pathways to provide specificity to activation of key developmental genes. Although much is known about transcription factors and target genes that regulate cardiomyocyte differentiation, less is known about scaffold proteins that couple signals at the cell surface to differentiation factors in developing heart cells. Here we show that AKAP13 (also known as Brx-1, AKAP-Lbc, and proto-Lbc), a unique protein kinase A-anchoring protein (AKAP) guanine nucleotide exchange region belonging to the Dbl family of oncogenes, is essential for cardiac development. Cardiomyocytes of Akap13-null mice had deficient sarcomere formation, and developing hearts were thin-walled and mice died at embryonic day 10.5-11.0. Disruption of Akap13 was accompanied by reduced expression of Mef2C. Consistent with a role of AKAP13 upstream of MEF2C, Akap13 siRNA led to a reduction in Mef2C mRNA, and overexpression of AKAP13 augmented MEF2C-dependent reporter activity. The results suggest that AKAP13 coordinates Galpha(12) and Rho signaling to an essential transcription program in developing cardiomyocytes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Fetal Heart/embryology , Fetal Heart/metabolism , Guanine Nucleotide Exchange Factors/metabolism , A Kinase Anchor Proteins/antagonists & inhibitors , A Kinase Anchor Proteins/deficiency , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Female , Fetal Heart/abnormalities , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , In Situ Hybridization , MEF2 Transcription Factors , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Minor Histocompatibility Antigens , Models, Cardiovascular , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Pregnancy , RNA, Small Interfering/genetics , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Signal Transduction , rho GTP-Binding Proteins/metabolism
14.
J Biol Chem ; 281(14): 9118-26, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16469733

ABSTRACT

Glucocorticoids regulate many crucial biologic functions through their cytoplasmic/nuclear glucocorticoid receptors (GR). Excess, deficiency, or alteration in tissue sensitivity to glucocorticoids has been associated with major causes of human morbidity and mortality. Brx, a cytoplasmic Rho family guanine nucleotide exchange factor, binds to and influences the activity of several nuclear hormone receptors. We examined the functional and molecular interactions between GR and Brx. The glucocorticoid sensitivity of lymphocytes obtained from mice haplo-insufficient for Brx was significantly decreased. Conversely, GR-mediated transcriptional activity of a glucocorticoid response element (GRE)-mediated glucocorticoid-responsive promoter was enhanced by Brx in a guanine nucleotide exchange factor domain-dependent fashion. Brx interacted with GR, forming a ternary complex with RhoA. In a chromatin immunoprecipitation assay, Brx and RhoA were co-precipitated with GREs only in the presence of ligand-activated GR. Extracellularly administered lysophosphatidic acid, which activates its signaling cascade through a specific membrane GTP-binding protein (G-protein)-coupled receptor in a G-protein alpha(13)-, Brx-, and RhoA-dependent fashion, enhanced GR transcriptional activity, whereas depletion of endogenous Brx attenuated this effect. These findings suggest that glucocorticoid signaling and, hence, the tissue sensitivity to glucocorticoids, may be coupled to extracellular signals via Brx and small G-proteins. Nuclear Brx might act as a local GRE-GR-transcriptosome activator by mediating the effect of small G-proteins on glucocorticoid-regulated genes.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Glucocorticoids/metabolism , Guanine Nucleotide Exchange Factors/physiology , Proto-Oncogene Proteins/physiology , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/physiology , Signal Transduction , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing/genetics , Animals , Chlorocebus aethiops , HeLa Cells , Humans , Kidney/cytology , Lymphocytes/physiology , Mice , Minor Histocompatibility Antigens , Monomeric GTP-Binding Proteins/metabolism , Plasmids , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Transfection , rho GTP-Binding Proteins/metabolism
15.
Biochem Biophys Res Commun ; 315(4): 1070-6, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14985122

ABSTRACT

Ski-interacting protein (SKIP), a vitamin D receptor (VDR) coactivator, also functions as a repressor in Notch signalling in association with the corepressor SMRT. Here we show that SKIP bifunctionally modulates (activates or represses) Retinoid-X receptor (RXR)- and VDR-dependent gene transcription in a cell line-specific manner, with activation in CV-1 and repression in P19 cells. The coactivator function of SKIP in these cells appeared to correlate with the relative level and ratio of expression of N-CoR and p300, with greater SKIP activation in higher p300-expressing and lower N-CoR-expressing cell-lines. C-terminal deletion of SKIP (delta334-536 aa) was associated with strong activation in both CV-1 and P19 cells. The corepressors N-CoR and SMRT and the coregulator p300 interacted with SKIP through the same N-terminal region (1-200 aa). Overall these results suggest that transcriptional action of SKIP may depend on distinct functional domains and cell line-specific interactions with both corepressors and coactivators.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Gene Expression , Histone Acetyltransferases , Mice , NIH 3T3 Cells , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 2 , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Retinoid X Receptors , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , p300-CBP Transcription Factors
16.
Trends Endocrinol Metab ; 13(10): 422-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12431838

ABSTRACT

In recent years, distinct signaling pathways involving specific complexes of cytoplasmic proteins have been shown to orchestrate estrogen action. These pathways might supplement or augment genomic effects of estrogen that are attributable to transcriptional activation by liganded receptor. Signals might be transduced through phosphorylation of the estrogen receptors (ERs), or indirectly through effects upon transcriptional coactivators or cell receptors. Estrogen signaling is coupled to growth factor signaling with feedback mechanisms directly impacting function of growth factor receptors. These signaling pathways regulate important physiological processes, such as cell growth and apoptosis. Here, we focus on cytoplasmic signaling pathways leading to activation of ERs.


Subject(s)
Estrogens/metabolism , Growth Substances/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/enzymology , Epidermal Growth Factor/metabolism , Feedback/physiology , Female , Mice , Protein Kinase C/metabolism , Receptor Cross-Talk , Transcriptional Activation/physiology , src Homology Domains
17.
Trends Endocrinol Metab ; 13(8): 349-54, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12217492

ABSTRACT

Remarkable progress in recent years has suggested that estrogen action in vivo is complex and often involves activation of cytoplasmic signaling cascades in addition to genomic actions mediated directly through estrogen receptors alpha and beta. Rather than a linear response mediated solely through estrogen-responsive DNA elements, in vivo estrogen might simultaneously activate distinct signaling cascades that function as networks to coordinate tissue responses to estrogen. This complex signaling system provides for exquisite control and plasticity of response to estrogen at the tissue level, and undoubtedly contributes to the remarkable tissue-specific responses to estrogens. In part I of this series, we summarize cytoplasmic signaling modules involving estrogen or estrogen receptors, with particular focus on recently described membrane-associated signaling complexes.


Subject(s)
Cell Membrane/metabolism , Cytoplasm/metabolism , Estrogens/pharmacology , Signal Transduction , Animals , Caveolin 1 , Caveolins/physiology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Mice , Mice, Knockout , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphatidylinositol 3-Kinases/physiology , Receptors, Estrogen/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...