Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Reprod Sci ; 24(1): 67-76, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27217374

ABSTRACT

The objective of the present study was to test the ability of OSU-03012 (2-amino-N-[4-[5-phenanthren-2-yl-3-(trifluoromethyl)pyrazol-1-yl]phenyl]acetamide), a novel and potent celecoxib-derivative, to impair endometriosis progression in in vitro and in vivo models based on its ability to indirectly block Y-box-binding protein 1 (YB-1) function. 12Z human endometriotic epithelial cells and sexually mature female C57BL/6J mice were treated with OSU-03012. Cellular proliferation was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromid assay. Expression of YB-1 and phosphorylated YB-1 in 12Z cells and endometriotic lesions was evaluated by Western blotting and immunohistochemistry (IHC). The IHC for proliferating cell nuclear antigen was performed. OSU-03012 treatment resulted in decreased YB-1 and its phosphorylated form in both in vitro and in vivo models. Endometriotic lesion size was significantly reduced in OSU-03012-treated mice (27.6 ± 4.0 mm3) compared to those from the control group (50.5 ± 6.9 mm3, P < .0001). A significant reduction in endometriotic epithelial cell proliferation was observed in endometriotic lesions exposed to OSU-03012 treatment ( P = .0346). In conclusion, targeting YB-1 via OSU-03012 showed a potent antiproliferative effect on endometriotic epithelial cells in vitro and in a mouse model of disease.

2.
Article in English | MEDLINE | ID: mdl-26779118

ABSTRACT

Endometriosis affects approximately 10% of women of reproductive age. This chronic, gynecological inflammatory disease results in a decreased quality of life for patients, with the main symptoms including chronic pelvic pain and infertility. The steroid hormone 17-ß Estradiol (E2) plays a key role in the pathology. Our previous studies showed that the anti-inflammatory lipid Lipoxin A4 (LXA4) acts as an estrogen receptor-alpha agonist in endometrial epithelial cells, inhibiting certain E2-mediated effects. LXA4 also prevents the progression of endometriosis in a mouse model via anti-proliferative mechanisms and by impacting mediators downstream of ER signaling. The aim of the present study was therefore to examine global proteomic changes evoked by E2 and LXA4 in endometriotic epithelial cells. E2 impacted a greater number of proteins in endometriotic epithelial cells than LXA4. Interestingly, the combination of E2 and LXA4 resulted in a reduced number of regulated proteins, with LXA4 mediating a suppressive effect on E2-mediated signaling. These proteins are involved in diverse pathways of relevance to endometriosis pathology and metabolism, including mRNA translation, growth, proliferation, proteolysis, and immune responses. In summary, this study sheds light on novel pathways involved in endometriosis pathology and further understanding of signaling pathways activated by estrogenic molecules in endometriotic epithelial cells.

3.
PLoS One ; 9(2): e89742, 2014.
Article in English | MEDLINE | ID: mdl-24587003

ABSTRACT

Endometriosis, a leading cause of pelvic pain and infertility, is characterized by ectopic growth of endometrial-like tissue and affects approximately 176 million women worldwide. The pathophysiology involves inflammatory and angiogenic mediators as well as estrogen-mediated signaling and novel, improved therapeutics targeting these pathways are necessary. The aim of this study was to investigate mechanisms leading to the establishment and progression of endometriosis as well as the effect of local treatment with Lipoxin A4 (LXA4), an anti-inflammatory and pro-resolving lipid mediator that we have recently characterized as an estrogen receptor agonist. LXA4 treatment significantly reduced endometriotic lesion size and downregulated the pro-inflammatory cytokines IL-1ß and IL-6, as well as the angiogenic factor VEGF. LXA4 also inhibited COX-2 expression in both endometriotic lesions and peritoneal fluid cells, resulting in attenuated peritoneal fluid Prostaglandin E2 (PGE2) levels. Besides its anti-inflammatory effects, LXA4 differentially regulated the expression and activity of the matrix remodeling enzyme matrix metalloproteinase (MMP)-9 as well as modulating transforming growth factor (TGF)-ß isoform expression within endometriotic lesions and in peritoneal fluid cells. We also report for first time that LXA4 attenuated aromatase expression, estrogen signaling and estrogen-regulated genes implicated in cellular proliferation in a mouse model of disease. These effects were observed both when LXA4 was administered prior to disease induction and during established disease. Collectively, our findings highlight potential targets for the treatment of endometriosis and suggest a pleotropic effect of LXA4 on disease progression, by attenuating pro-inflammatory and angiogenic mediators, matrix remodeling enzymes, estrogen metabolism and signaling, as well as downstream proliferative pathways.


Subject(s)
Biosynthetic Pathways/drug effects , Dinoprostone/biosynthesis , Endometriosis/prevention & control , Estrogens/metabolism , Lipoxins/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Aromatase/metabolism , Cyclooxygenase 2/metabolism , DNA Primers/genetics , Endometriosis/physiopathology , Female , Immunohistochemistry , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
4.
PLoS One ; 8(12): e82512, 2013.
Article in English | MEDLINE | ID: mdl-24324802

ABSTRACT

BACKGROUND/AIMS: The neural cell adhesion molecule L1CAM is a transmembrane glycoprotein abnormally expressed in tumors and previously associated with cell proliferation, adhesion and invasion, as well as neurite outgrowth in endometriosis. Being an attractive target molecule for antibody-based therapy, the present study assessed the ability of the monoclonal anti-L1 antibody (anti-L1 mAb) to impair the development of endometriotic lesions in vivo and endometriosis-associated nerve fiber growth. METHODS AND RESULTS: Endometriosis was experimentally induced in sexually mature B6C3F1 (n=34) and CD-1 nude (n=21) mice by autologous and heterologous transplantation, respectively, of endometrial fragments into the peritoneal cavity. Transplantation was confirmed four weeks post-surgery by in vivo magnetic resonance imaging and laparotomy, respectively. Mice were then intraperitoneally injected with anti-L1 mAb or an IgG isotype control antibody twice weekly, over a period of four weeks. Upon treatment completion, mice were sacrificed and endometrial implants were excised, measured and fixed. Endometriosis was histologically confirmed and L1CAM was detected by immunohistochemistry. Endometriotic lesion size was significantly reduced in anti-L1-treated B6C3F1 and CD-1 nude mice compared to mice treated with control antibody (P<0.05). Accordingly, a decreased number of PCNA positive epithelial and stromal cells was detected in autologously and heterologously induced endometriotic lesions exposed to anti-L1 mAb treatment. Anti-L1-treated mice also presented a diminished number of intraperitoneal adhesions at implantation sites compared with controls. Furthermore, a double-blind counting of anti-neurofilament L stained nerves revealed significantly reduced nerve density within peritoneal lesions in anti-L1 treated B6C3F1 mice (P=0.0039). CONCLUSIONS: Local anti-L1 mAb treatment suppressed endometriosis growth in B6C3F1 and CD-1 nude mice and exerted a potent anti-neurogenic effect on induced endometriotic lesions in vivo. The findings of this preliminary study in mice provide a strong basis for further testing in in vivo models.


Subject(s)
Antibodies, Monoclonal/pharmacology , Endometriosis/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Adult , Animals , Antibodies, Monoclonal/administration & dosage , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endometriosis/drug therapy , Endometriosis/pathology , Female , Humans , Mice , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neurofilament Proteins/metabolism , Young Adult
5.
Fertil Steril ; 99(7): 1965-73.e2, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23472950

ABSTRACT

OBJECTIVE: To compare the expression of the prostaglandin (PG) E(2) transporter multidrug resistance-associated protein 4 (MRP4) in eutopic and ectopic endometrial tissue from endometriosis patients with that of control subjects and to examine whether MRP4 is regulated by the antiinflammatory lipid lipoxin A(4) (LXA(4)) in endometriotic epithelial cells. DESIGN: Molecular analysis in human samples and a cell line. SETTING: Two university hospitals and a private clinic. PATIENT(S): A total of 59 endometriosis patients and 32 age- and body mass index-matched control subjects undergoing laparoscopy or hysterectomy. INTERVENTION(S): Normal, eutopic, and ectopic endometrial biopsies as well as peritoneal fluid were obtained during surgery performed during the proliferative phase of the menstrual cycle. 12Z endometriotic epithelial cells were used for in vitro mechanistic studies. MAIN OUTCOME MEASURE(S): Tissue MRP4 mRNA levels were quantified by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and localization was analyzed with the use of immunohistochemistry. Cellular MRP4 mRNA and protein were quantified by qRT-PCR and Western blot, respectively. PGE(2) was measured in peritoneal fluid and cell supernatants using an enzyme immunoassay (EIA). RESULT(S): MRP4 was expressed in eutopic and ectopic endometrium, where it was overexpressed in peritoneal lesions and localized in the cytoplasm of glandular epithelial cells. LXA(4) attenuated MRP4 mRNA and protein levels in endometriotic epithelial cells in a dose-dependent manner, while not affecting the expression of enzymes involved in PGE(2) metabolism. Investigations employing receptor antagonists and small interfering RNA revealed that this occurred through estrogen receptor α. Accordingly, LXA(4) treatment inhibited extracellular PGE(2) release. CONCLUSION(S): We report for the first time that MRP4 is expressed in human endometrium, elevated in peritoneal endometriosis, and modulated by LXA(4) in endometriotic epithelial cells.


Subject(s)
Endometriosis/metabolism , Endometrium/metabolism , Epithelial Cells/metabolism , Lipoxins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Peritoneal Diseases/metabolism , Adult , Ascitic Fluid/metabolism , Biopsy , Blotting, Western , Case-Control Studies , Cell Line , Dinoprostone/metabolism , Endometriosis/genetics , Endometriosis/surgery , Endometrium/drug effects , Endometrium/surgery , Epithelial Cells/drug effects , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Hysterectomy , Immunoenzyme Techniques , Immunohistochemistry , Laparoscopy , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Peritoneal Diseases/genetics , Peritoneal Diseases/surgery , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation , Young Adult
6.
Fertil Steril ; 98(5): 1200-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22884659

ABSTRACT

OBJECTIVE: To analyze the expression of estrogen receptors α and ß as well as their target genes implicated in proliferation, c-myc, cyclin D1, and GREB1, in the endometrium of women with or without endometriosis. DESIGN: Expression analysis in human tissue. SETTING: University hospitals and a clinic. PATIENT(S): Ninety-one premenopausal women (59 patients with endometriosis and 32 controls) undergoing laparoscopic surgery. INTERVENTION(S): Biopsies were obtained at time of surgery, performed during the proliferative phase of the cycle. MAIN OUTCOME MEASURE(S): Estrogen receptors α and ß as well as c-myc, cyclin D1, and GREB1 mRNA expression levels were determined by quantitative reverse transcriptase-polymerase chain reaction. Tissue localization of these estrogen-regulated genes was analyzed by immunohistochemistry. RESULT(S): Estrogen receptors α and ß as well as c-myc, cyclin D1, and GREB1 mRNA expression levels were increased in ectopic tissue in comparison with both normal and eutopic endometrium. Estrogen receptor mRNA levels also were upregulated in the eutopic peritoneal tissue of patients with endometriosis. Cyclin D1 and GREB1 expression was augmented in eutopic endometrium. c-myc, cyclin D1, and GREB1 proteins exhibited a nuclear localization in ectopic endometrial tissue. CONCLUSION(S): This constitutes the first report of increased expression of GREB1, as well as cyclin D1 and c-myc, in peritoneal endometriotic lesions, implicating these proteins in estrogen-dependent growth in this context.


Subject(s)
Cell Proliferation , Choristoma/metabolism , Cyclin D1/analysis , Endometriosis/metabolism , Endometrium , Estrogen Receptor alpha/analysis , Estrogen Receptor beta/analysis , Neoplasm Proteins/analysis , Peritoneal Diseases/metabolism , Proto-Oncogene Proteins c-myc/analysis , Adult , Biopsy , Case-Control Studies , Choristoma/genetics , Choristoma/pathology , Choristoma/surgery , Cyclin D1/genetics , Endometriosis/genetics , Endometriosis/pathology , Endometriosis/surgery , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation , Germany , Humans , Immunohistochemistry , Middle Aged , Neoplasm Proteins/genetics , Peritoneal Diseases/genetics , Peritoneal Diseases/pathology , Peritoneal Diseases/surgery , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Switzerland , Young Adult
7.
FASEB J ; 25(12): 4326-37, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21885654

ABSTRACT

Inflammation is intimately linked with naturally occurring remodeling events in the endometrium. Lipoxins comprise a group of short-lived, nonclassic eicosanoids possessing potent anti-inflammatory and proresolution properties. In the present study, we investigated the role of lipoxin A(4) (LXA(4)) in the endometrium and demonstrated that 15-LOX-2, an enzyme necessary for LX biosynthesis, is expressed in this tissue. Our results establish that LXA(4) possesses robust estrogenic activity through its capacity to alter ERE transcriptional activity, as well as expression of estrogen-regulated genes, alkaline phosphatase activity, and proliferation in human endometrial epithelial cells. Interestingly, LXA(4) also demonstrated antiestrogenic potential, significantly attenuating E2-induced activity. This estrogenic activity was directly mediated through estrogen receptors (ERs). Subsequent investigations determined that the actions of LXA(4) are exclusively mediated through ERα and closely mimic those of the potent estrogen 17ß-estradiol (E2). In binding assays, LXA(4) competed with E2 for ER binding, with an IC(50) of 46 nM. Furthermore, LXA(4) exhibited estrogenic activity in vivo, increasing uterine wet weight and modulating E2-regulated gene expression. These findings reveal a previously unappreciated facet of LXA(4) bioactions, implicating this lipid mediator in novel immunoendocrine crosstalk mechanisms.


Subject(s)
Estrogen Receptor Modulators/metabolism , Lipoxins/metabolism , Alkaline Phosphatase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Base Sequence , Binding, Competitive , Cell Line , Cell Proliferation/drug effects , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/metabolism , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Lipoxins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/antagonists & inhibitors , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Signal Transduction
8.
J Reprod Immunol ; 92(1-2): 88-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21940052

ABSTRACT

Immunity and hormonal responses in the reproductive tissues of postmenopausal women are poorly understood. Secretory leukocyte protease inhibitor (SLPI), a multifunctional antimicrobial protein expressed at mucosal surfaces, is thought to play a key role in infectious and inflammatory contexts. The aim of this study was to measure SLPI production along the female reproductive tract in postmenopausal women with and without hormonal treatment. We additionally quantified estrogen receptor alpha (ERα) and progesterone receptor A (PRA) in these tissues. Expression of SLPI was decreased in the vagina and ectocervix of women under hormonal treatment. Endocervical ERα mRNA expression was increased while this did not reach significance at the protein level. SLPI expression in the endometrium was not influenced by hormonal treatment. We observed attenuated ERα expression in the cervix and endometrium of hormonally treated women, whereas vaginal expression was increased. PRA expression was augmented in the cervix and endometrium and unchanged in the vagina. Taken together, our results indicate that hormonal responses and receptor expression are differentially regulated in vaginal tissue compared with the cervix and endometrium.


Subject(s)
Genitalia, Female/metabolism , Hormones/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Aged , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Replacement Therapy , Female , Gene Expression Profiling , Gene Expression Regulation , Genitalia, Female/immunology , Genitalia, Female/pathology , Humans , Immunity, Mucosal , Middle Aged , Postmenopause , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Secretory Leukocyte Peptidase Inhibitor/genetics
9.
Mol Cell Endocrinol ; 345(1-2): 27-37, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21784129

ABSTRACT

The pro-inflammatory cytokine TNF-α and the female hormone estrogen have been implicated in the pathophysiology of two common gynecological diseases, endometriosis and endometrial adenocarcinoma. Here we describe a novel capacity of TNF-α to activate ER signaling in endometrial epithelial cells. TNF-α induced luciferase expression in the absence and presence of estradiol and also augmented expression of the estrogen-regulated genes c-fos, GREB1, and progesterone receptor. Furthermore, TNF-α mediated ER transcriptional activity is dependent on the Extracellular Regulated Kinase (ERK) 1/2 pathway. Co-treatment with a pure ER antagonist resulted in an inhibition of this TNF-α-induced ERE luciferase activity and gene expression, demonstrating that this cytokine signals through ERs. Additional investigations confirmed that TNF-α acts specifically via ERα. Taken together, these data provide a rationale for the potential use of inhibitors of TNF-α and estrogen production/activity in combination for the treatment of endometrial pathologies.


Subject(s)
Endometrium/cytology , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Luciferases/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptor Cross-Talk/drug effects , Response Elements/genetics , Signal Transduction/genetics , Transcription, Genetic
11.
FASEB J ; 21(12): 3162-70, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17496159

ABSTRACT

Migration of neutrophils (PMN) across epithelia is a pathological hallmark of numerous mucosal diseases. Whereas lesions at mucosal surfaces are generally self-limiting, endogenous mechanisms of resolution are incompletely understood. Previous studies revealed that resolvins directly act on PMN to attenuate transendothelial migration, less is known about the influence of resolvins on PMN-epithelial interactions and whether they act on epithelia. We studied the dynamics of resolvin E1 (RvE1) actions on leukocyte transepithelial migration. PMN exposure to RvE1 or chemerin (peptide agonist of ChemR23) reduced transepithelial migration in a concentration-dependent manner. Conversely, activation of epithelial ChemR23 promoted apical clearance of PMN. A nonbiased screen of known PMN ligands expressed on epithelial cells in response to RvE1 revealed selective induction of CD55, an apically expressed antiadhesive molecule. CD55 promoter analysis demonstrated that both RvE1 and chemerin activate the CD55 promoter. Inhibition of CD55 by neutralizing antibody prevented RvE1-dependent augmentation of apical PMN clearance. Taken together these findings implicate a "two-hit" model of inflammatory resolution, whereby activation of the PMN RvE1 receptor attenuates transepithelial migration and subsequent actions on the epithelium promote CD55-dependent clearance of PMN across the epithelial cell surface promoting active inflammatory resolution.


Subject(s)
Cell Movement/physiology , Eicosapentaenoic Acid/analogs & derivatives , Epithelial Cells , Inflammation/immunology , Neutrophils/immunology , Animals , CD55 Antigens/genetics , CD55 Antigens/metabolism , Cell Line , Chemokines/metabolism , Eicosapentaenoic Acid/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/physiology , Humans , Intercellular Adhesion Molecule-1/metabolism , Intercellular Signaling Peptides and Proteins , Mucous Membrane/cytology , Mucous Membrane/immunology , Neutrophils/cytology , Neutrophils/physiology , Promoter Regions, Genetic
12.
J Infect Dis ; 194(4): 498-502, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16845634

ABSTRACT

Genital tract epithelia regularly encounter and adapt to the existence of bacterial pathogens. This study provides evidence that the endocervical and ectocervical epithelia of the human female genital tract express bactericidal/permeability-increasing protein (BPI). The constitutive expression of BPI was restricted to cell-bound protein and unaffected by human papillomavirus type 16/E6E7 immortalization and proinflammatory cytokine stimulation. Epithelial BPI was, in part, responsible for killing a commensal strain of Escherichia coli. The results of the present study suggest that BPI is tightly regulated and functionally expressed by epithelial cells in the female reproductive tract and may play a role in regulating bacterial colonization in the genital mucosa.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Cervix Uteri/cytology , Epithelial Cells/metabolism , Membrane Proteins/metabolism , Antimicrobial Cationic Peptides/pharmacology , Blood Proteins/pharmacology , Cells, Cultured , Escherichia coli/drug effects , Female , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Membrane Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...