Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
medRxiv ; 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38045369

ABSTRACT

The fallopian tube, connecting the uterus with the ovary, is a dynamic organ that undergoes cyclical changes and is the site of several diseases, including serous cancer. Here, we use single-cell technologies to construct a comprehensive cell map of healthy pre-menopausal fallopian tubes, capturing the impact of the menstrual cycle and menopause on different fallopian tube cells at the molecular level. The comparative analysis between pre- and post-menopausal fallopian tubes reveals substantial shifts in cellular abundance and gene expression patterns, highlighting the physiological changes associated with menopause. Further investigations into menstrual cycle phases illuminate distinct molecular states in secretory epithelial cells caused by hormonal fluctuations. The markers we identified characterizing secretory epithelial cells provide a valuable tool for classifying ovarian cancer subtypes. Graphical summary: Graphical summary of results. During the proliferative phase (estrogen high ) of the menstrual cycle, SE2 cells (OVGP1 + ) dominate the fallopian tube (FT) epithelium, while SE1 cells (OVGP1 - ) dominate the epithelium during the secretory phase. Though estrogen levels decrease during menopause, SE post-cells (OVGP1 + , CXCL2 + ) make up most of the FT epithelium.

2.
Nat Commun ; 7: 12645, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561551

ABSTRACT

A cell line representative of human high-grade serous ovarian cancer (HGSOC) should not only resemble its tumour of origin at the molecular level, but also demonstrate functional utility in pre-clinical investigations. Here, we report the integrated proteomic analysis of 26 ovarian cancer cell lines, HGSOC tumours, immortalized ovarian surface epithelial cells and fallopian tube epithelial cells via a single-run mass spectrometric workflow. The in-depth quantification of >10,000 proteins results in three distinct cell line categories: epithelial (group I), clear cell (group II) and mesenchymal (group III). We identify a 67-protein cell line signature, which separates our entire proteomic data set, as well as a confirmatory publicly available CPTAC/TCGA tumour proteome data set, into a predominantly epithelial and mesenchymal HGSOC tumour cluster. This proteomics-based epithelial/mesenchymal stratification of cell lines and human tumours indicates a possible origin of HGSOC either from the fallopian tube or from the ovarian surface epithelium.


Subject(s)
Epithelial Cells/pathology , Gene Expression Profiling , Ovarian Neoplasms/pathology , Proteomics/methods , Cell Line, Tumor , Datasets as Topic , Fallopian Tubes/cytology , Fallopian Tubes/pathology , Female , Humans , Mass Spectrometry/methods , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovary/cytology , Ovary/pathology , Primary Cell Culture , Transcriptome
3.
Oncogene ; 34(48): 5857-68, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-25867073

ABSTRACT

Cancer cells grow in an environment comprised of multiple components that support tumor growth and contribute to therapy resistance. Major cell types in the tumor microenvironment are fibroblasts, endothelial cells and infiltrating immune cells all of which communicate with cancer cells. One way that these cell types promote cancer progression is by altering the expression of microRNAs (miRNAs), small noncoding RNAs that negatively regulate protein expression, either in the cancer cells or in the associated normal cells. Changes in miRNA expression can be brought about by direct interaction between the stromal cells and cancer cells, by paracrine factors secreted by any of the cell types or even through direct communication between cells through secreted miRNAs. Understanding the role of miRNAs in the complex interactions between the tumor and cells in its microenvironment is necessary if we are to understand tumor progression and devise new treatments.


Subject(s)
Cell Communication/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment/genetics , Animals , Humans
4.
Oncogene ; 34(48): 5923-32, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-25798837

ABSTRACT

The cross-talk between ovarian cancer (OvCa) cells and the metastatic microenvironment is an essential determinant of successful colonization. MicroRNAs (miRNAs) have several critical roles during metastasis; however, the role of microenvironmental cues in the regulation of miRNAs in metastasizing cancer cells has not been studied. Using a three-dimensional culture model that mimics the human omentum, one of the principal sites of OvCa metastasis, we identified and characterized the microenvironment-induced downregulation of a tumor suppressor miRNA, miR-193b, in metastasizing OvCa cells. The direct interaction of the OvCa cells with mesothelial cells, which cover the surface of the omentum, caused a DNA methyltransferase 1-mediated decrease in the expression of miR-193b in the cancer cells. The reduction in miR-193b enabled the metastasizing cancer cells to invade and proliferate into human omental pieces ex vivo and into the omentum of a mouse xenograft model of OvCa metastasis. The functional effects of miR-193b were mediated, in large part, by the concomitant increased expression of its target, urokinase-type plasminogen activator, a known tumor-associated protease. These findings link paracrine signals from the microenvironment to the regulation of a key miRNA in cancer cells. Targeting miR-193b, which is essential for metastatic colonization of cancer cells could prove effective in the treatment of OvCa metastasis.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Omentum/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Tumor Microenvironment , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Proliferation , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Omentum/metabolism , Ovarian Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Xenograft Model Antitumor Assays
5.
Oncogene ; 34(26): 3452-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25174399

ABSTRACT

We previously characterized the link between WNT7A and the progression of ovarian cancer. Other groups have identified FGF1 as a relevant risk factor in ovarian cancer. Here, we show a linkage between these two signaling pathways that may be exploited to improve treatment and prognosis of patients with ovarian cancer. High expression of WNT7A and FGF1 are correlated in ovarian carcinomas and poor overall patient survival. A chromatin immunoprecipitation assay demonstrated that WNT7A/ß-catenin signaling directly regulates FGF1 expression via TCF binding elements in the FGF1-1C promoter locus. In vitro gene manipulation studies revealed that FGF1 is sufficient to drive the tumor-promoting effects of WNT7A. In vivo xenograft studies confirmed that the stable overexpression of WNT7A or FGF1 induced a significant increase in tumor incidence, whereas FGF1 knockdown in WNT7A overexpressing cells caused a significant reduction in tumor size. Niclosamide most efficiently abrogated WNT7A/ß-catenin signaling in our model, inhibited ß-catenin transcriptional activity and cell viability, and increased cell death. Furthermore, niclosamide decreased cell migration following an increase in E-cadherin subsequent to decreased levels of SLUG. The effects of niclosamide on cell functions were more potent in WNT7A-overexpressing cells. Oral niclosamide inhibited tumor growth and progression in an intraperitoneal xenograft mouse model representative of human ovarian cancer. Collectively, these results indicate that FGF1 is a direct downstream target of WNT7A/ß-catenin signaling and this pathway has potential as a therapeutic target in ovarian cancer. Moreover, niclosamide is a promising inhibitor of this pathway and may have clinical relevance.


Subject(s)
Drug Resistance, Neoplasm , Fibroblast Growth Factor 1/genetics , Niclosamide/pharmacology , Ovarian Neoplasms , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Fibroblast Growth Factor 1/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Wnt Signaling Pathway/physiology
6.
Oncogene ; 33(28): 3619-33, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-23934194

ABSTRACT

Epithelial ovarian cancer (OvCa) is associated with high mortality and, as the majority (>75%) of women with OvCa have metastatic disease at the time of diagnosis, rates of survival have not changed appreciably over 30 years. A mechanistic understanding of OvCa initiation and progression is hindered by the complexity of genetic and/or environmental initiating events and lack of clarity regarding the cell(s) or tissue(s) of origin. Metastasis of OvCa involves direct extension or exfoliation of cells and cellular aggregates into the peritoneal cavity, survival of matrix-detached cells in a complex ascites fluid phase and subsequent adhesion to the mesothelium lining covering abdominal organs to establish secondary lesions containing host stromal and inflammatory components. Development of experimental models to recapitulate this unique mechanism of metastasis presents a remarkable scientific challenge, and many approaches used to study other solid tumors (for example, lung, colon and breast) are not transferable to OvCa research given the distinct metastasis pattern and unique tumor microenvironment (TME). This review will discuss recent progress in the development and refinement of experimental models to study OvCa. Novel cellular, three-dimensional organotypic, and ex vivo models are considered and the current in vivo models summarized. The review critically evaluates currently available genetic mouse models of OvCa, the emergence of xenopatients and the utility of the hen model to study OvCa prevention, tumorigenesis, metastasis and chemoresistance. As these new approaches more accurately recapitulate the complex TME, it is predicted that new opportunities for enhanced understanding of disease progression, metastasis and therapeutic response will emerge.


Subject(s)
Models, Biological , Neoplasms, Glandular and Epithelial , Ovarian Neoplasms , Animals , Animals, Genetically Modified , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Tumor Microenvironment
7.
Phys Rev Lett ; 107(5): 057001, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21867090

ABSTRACT

We carried out specific-heat and ac-susceptibility experiments under hydrostatic pressure to investigate the interplay of spin-density-wave antiferromagnetism (A) and superconductivity (S) in single-crystalline AS-type CeCu(2)Si(2). We find evidence for a line of magnetic-field- and pressure-tuned quantum critical points in the normal state in the zero-temperature magnetic field-pressure plane. Our analysis suggests an extension of this line into the superconducting state and corroborates the close connection of the underlying mechanisms leading to the formation of the antiferromagnetic and the superconducting states in AS-type CeCu(2)Si(2).

8.
Oncogene ; 30(13): 1566-76, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21119598

ABSTRACT

The role of the fibronectin receptor, α(5)ß(1)-integrin, as an adhesion receptor and in angiogenesis is well established. However, its role in cancer cell invasion and metastasis is less clear. We describe a novel mechanism by which fibronectin regulates ovarian cancer cell signaling and promotes metastasis. Fibronectin binding to α(5)ß(1)-integrin led to a direct association of α(5)-integrin with the receptor tyrosine kinase, c-Met, activating it in a hepatocyte growth factor/scatter factor (HGF/SF) independent manner. Subsequently, c-Met associated with Src, and activated Src and focal adhesion kinase (FAK). Inhibition of α(5)ß(1)-integrin decreased the phosphorylation of c-Met, FAK and Src, both in vitro and in vivo. Independent activation of c-Met by its native ligand, HGF/SF, or overexpression of a constitutively active FAK in HeyA8 cells could overcome the effect of α(5)ß(1)-integrin inhibition on tumor cell invasion, indicating that α(5)ß(1)-integrin is upstream of c-Met, Src and FAK. Inhibition of α(5)ß(1)-integrin on cancer cells in two xenograft models of ovarian cancer metastasis resulted in a significant decrease of tumor burden, which was independent of the effect of α(5)ß(1)-integrin on angiogenesis. These data suggest that fibronectin promotes ovarian cancer invasion and metastasis through an α(5)ß(1)-integrin/c-Met/FAK/Src-dependent signaling pathway, transducing signals through c-Met in an HGF/SF-independent manner.


Subject(s)
Fibronectins/physiology , Integrin alpha5beta1/physiology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-met/physiology , Animals , Cell Line, Tumor , Female , Focal Adhesion Kinase 1/metabolism , Humans , Ligands , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/etiology , Ovarian Neoplasms/blood supply , Phosphorylation , Signal Transduction , src-Family Kinases/metabolism
9.
Gynecol Oncol ; 111(3): 537-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18554701

ABSTRACT

BACKGROUND: Reversible posterior leukoencephalopathy syndrome (RPLS) is a clinical and radiologic syndrome of heterogeneous etiology. Although it has been described in association with intravenous cytotoxic chemotherapy including cisplatin and molecularly targeted therapies such as bevacizumab and sorafenib, it has not been described in the setting of intraperitoneal chemotherapy. CASE: A 64-year-old woman with stage IIIC fallopian tube cancer developed acute mental status changes and radiologic findings consistent with RPLS in conjunction with hypertension after one cycle of intravenous paclitaxel, followed by intraperitoneal (IP) cisplatin. Symptoms resolved over the course of 4 days with no obvious residual effects. CONCLUSION: The use of an intravenous paclitaxel and intraperitoneal cisplatin regimen can be associated with RPLS. Hypertension should be controlled in patients prior to receiving this chemotherapy regimen due to potential toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Fallopian Tube Neoplasms/drug therapy , Posterior Leukoencephalopathy Syndrome/chemically induced , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/administration & dosage , Cisplatin/adverse effects , Female , Humans , Infusions, Intravenous , Infusions, Parenteral , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects
10.
Histopathology ; 51(1): 54-62, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17593080

ABSTRACT

AIMS: Tyrosine kinase receptors Her2/neu and c-Met play an important role in breast cancer development and progression. Our aim was to determine the expression of c-Met, its ligand hepatocyte growth factor/scatter factor (HGF/SF) and Her2/neu in ductal carcinoma in situ (DCIS) lesions of the breast (n = 39) by two different immunocytochemical techniques, classical immunohistochemistry and immunofluorescence, and to correlate their expression levels with histopathological and clinical characteristics. METHODS AND RESULTS: Both methods revealed similar c-Met staining patterns in both the in situ component and the adjacent normal tissue (P < 0.001). However, an imbalance in c-Met expression between tumour and surrounding normal tissue was correlated with high-grade DCIS (Van Nuys Grade 3). No correlation existed between Her2/neu and c-Met expression. High HGF/SF immunoreactivity was observed in 43.6% of the cases, yet the adjacent cellular stroma revealed only low levels of HGF/SF. No correlation existed between c-Met, Her2/neu or HGF/SF expression and clinicopathological factors. CONCLUSION: An imbalance in c-Met expression between tumour and surrounding normal tissue is associated with an aggressive DCIS phenotype. Moreover, c-Met and HGF/SF may contribute to tumour development by different means than those controlled by Her2/neu.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Breast/cytology , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Humans , Middle Aged , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics
11.
Gynecol Oncol ; 104(3): 524-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17049587

ABSTRACT

OBJECTIVE: Uterine sarcomas are a heterogeneous group of tumors with a propensity for metastasis and resistance to conventional therapy. Recent success in the treatment of other solid tumors with the targeted tyrosine kinase inhibitor imatinib mesylate offers new avenues for investigation. The primary target of imatinib is c-kit, but the drug also inhibits PDGFR-alpha and PDGFR-beta. Given the lack of identified molecular targets in endometrial stromal sarcomas, leiomyosarcomas, and carcinosarcomas, the purpose of this study was to determine the protein expression of c-kit, PDGFR-alpha, and PDGFR-beta in these tumors as a preliminary step to determining their susceptibility to directed therapy. A secondary goal was to identify specific gene mutations that might be associated with activation of these proteins in uterine sarcomas. METHODS: Archived tissue from 42 cases of uterine sarcomas was stained for c-kit, PDGFR-alpha, and PDGFR-beta using immunohistochemistry. Laser-capture microdissected samples of uterine carcinosarcomas, or homogeneous areas of leiomyosarcomas or endometrial stromal sarcomas, were subjected to genetic analysis of PDGFR-alpha exons 12 and 18. RESULTS: The majority (38/42, 90%) of uterine sarcomas lacked c-kit expression and 90% (38/42) demonstrated negative or weak staining for PDGFR-beta. In contrast, 70% (30/42) of cases had strong staining for PDGFR-alpha in the tumor but not in normal myometrium or endometrium. Sequencing results revealed no mutations in exons 12 or 18 of PDGFR-alpha. CONCLUSION: c-kit and PDGFR-beta are unlikely to represent primary treatment targets in uterine sarcomas. The strong expression of PDGFR-alpha in uterine sarcoma specimens suggests a role for this receptor in tumor development, although its potential as a therapeutic target requires further investigation.


Subject(s)
Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Sarcoma/enzymology , Uterine Neoplasms/enzymology , Female , Humans , Proto-Oncogene Proteins c-kit/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Sarcoma/pathology , Uterine Neoplasms/pathology
12.
Eur J Surg Oncol ; 31(2): 183-90, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15698736

ABSTRACT

AIM: To report the role of liver angiography in the staging of medullary thyroid cancer (MTC) patients. MATERIAL AND METHODS: Sixty MTC patients with persistent or recurrent hypercalcitonemia (n=49), a characteristic general symptom (diarrhea, n=4) or a normal basal calcitonin level without general symptoms (n=7) were investigated by dynamic liver CT, MRI and angiography between 06/1998 and 06/2002. RESULTS: Dual-phase CT and MRI investigations identified hepatic metastases with relatively low frequency (8/58 on MRI, and 7/60 on CT). Angiography indicated liver involvement in 54/60 cases. The hepatic metastases were typically multiple, hypervascular, small foci (only 13 foci measured >/=10 mm). With one exception significant disease progression was not observed over 5 years of follow-up. CONCLUSIONS: Liver angiography is a powerful tool to reveal hepatic metastases in MTC patients. Frequent, inoperable liver metastases in hypercalcitoninemic MTC patients demonstrate that secondary lymph node dissection is an inefficient technique for restoration of a normal calcitonin level.


Subject(s)
Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/surgery , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Liver/pathology , Liver/surgery , Lymph Node Excision , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/surgery , Angiography , Biomarkers, Tumor/blood , Brain Stem Neoplasms/blood , Calcitonin/blood , Cervix Uteri/metabolism , Cervix Uteri/pathology , Cervix Uteri/surgery , Female , Follow-Up Studies , Humans , Liver/metabolism , Liver Neoplasms/blood , Lung Neoplasms/blood , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Lymphatic Metastasis , Magnetic Resonance Imaging , Male , Mediastinum/pathology , Mediastinum/surgery , Neoplasms, Bone Tissue/blood , Neoplasms, Bone Tissue/secondary , Neoplasms, Bone Tissue/surgery , Thyroid Neoplasms/blood , Tomography, X-Ray Computed , Treatment Outcome
13.
Spinal Cord ; 40(9): 468-73, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185608

ABSTRACT

OBJECTIVE: To investigate the pathomechanism of the rare radiogenic lower motor neurone disease (LMND) on the basis of a case history involving a partial functional recovery. PATIENT: A 31-year-old seminoma patient received postoperative para-aortic and para-iliac telecobalt irradiation with a biologically effective dose of 88 Gy(2) (44 Gy in 2 Gy fractions/day, with an estimated alpha/beta of 2 Gy) delivered to the spinal cord following a single cycle of chemotherapy. LMND developed 4 months after the completion of radiotherapy. The patient exhibited flaccid paraparesis of the lower extremities (without sensory or vegetative signs), followed by a worsening after further chemotherapy, due to pulmonary metastatization. A gradual spontaneous functional improvement commenced and led several years later to a stabilized state involving moderately severe symptoms. METHODS: In the 15th year of the clinical course, magnetic resonance imaging (MRI) and positron emission tomography (PET) with [(18)F]fluorodeoxyglucose (FDG) and [(11)C] methionine were conducted. Four lines of experiments (clonogenic assay using fibroblasts isolated from a skin biopsy sample of the patient, comet assay, micronucleus assay, and the testing of chromosome aberrations after in vitro irradiation of peripheral blood samples) were performed in a search for an increased individual radiosensitivity. RESULTS: MRI investigations failed to reveal any pathological change. PET demonstrated an increased FDG accumulation, but a negligible [(11)C] methionine uptake in the irradiated spinal cord segments. The radiobiological investigations did not indicate any sign of an increased individual radiosensitivity. CONCLUSIONS: We suggest that the observed partial functional recovery and stabilization of the symptoms of radiogenic LMND may be explained by the higher than normal density of sodium channels expressed along the demyelinated axons of the restored conduction. The increased energy demands of this type of conduction are proved by a higher metabolic rate (increased FDG uptake) of the irradiated spinal cord segments without a substantial regenerative process (lack of detectable protein synthesis).


Subject(s)
Cobalt Radioisotopes/adverse effects , Motor Neuron Disease/etiology , Motor Neuron Disease/physiopathology , Radioisotope Teletherapy/adverse effects , Tomography, Emission-Computed , Adult , Fluorodeoxyglucose F18 , Humans , Magnetic Resonance Imaging , Male , Motor Neuron Disease/diagnostic imaging , Motor Neuron Disease/metabolism , Radiopharmaceuticals , Recovery of Function , Seminoma/therapy , Sodium Channels/metabolism , Testicular Neoplasms/therapy , Tomography, Emission-Computed/methods
14.
Gynecol Oncol ; 82(2): 291-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11531282

ABSTRACT

OBJECTIVE: Matrix metalloproteinases (MMPs) are frequently expressed in malignant tumors and play an important role in tumor invasion and metastasis. MMP-2 and MMP-9 expression has been correlated with poor survival in some tumors, but data for ovarian cancer are lacking, despite clinical trials with MMP inhibitors. The aim of this study was to assess activity of MMP-2 and MMP-9 and correlate it to prognosis in ovarian cancer. METHODS: MMP-2 and MMP-9 gelatinolytic activity was analyzed in 84 patients with advanced ovarian cancer FIGO stage III and 19 benign ovarian tumors by gelatin zymography. MMP-9 immunoreactivity was detected by immunohistochemistry and gelatinolytic activity was localized in ovarian cancer tissue by in situ zymography. RESULTS: were correlated with patient survival, with a median follow-up period of 55 months. Results. Median pro-MMP-9 activity was at 0.00 U/microg protein in benign ovarian tissues and 4.82 U/microg protein in ovarian cancer (P = 0.001); activated MMP-9 was not detected. Pro-MMP-2 expression in benign ovarian tissue did not differ from that of malignant ovarian tissue, whereas active MMP-2 was present in 52% of ovarian cancers, but absent in benign ovarian tissues. Analyzing all patients high pro-MMP-9 activity was associated with short overall survival (P = 0.019) while pro-MMP-2 and activated MMP-2 did not predict overall survival. When analyzing the subgroups of patients with and without residual tumor mass at the time of surgery, pro-MMP-9 was of prognostic value only in the subgroup of patients with no residual tumor mass. In univariate analysis pro-MMP-9 activity, residual tumor mass, age, ascites volume, and grading were of prognostic significance for overall survival. However, in multivariate analyses, including all biological and clinicopathologic variables, only pro-MMP-9 and residual disease remained statistically independent prognostic factors. In situ zymography localized gelatinolytic activity predominantly to the tumor cell nests displaying MMP-9 immunoreactivity. CONCLUSIONS: Pro-MMP-9 gelatinolytic activity, but not active MMP-2 or MMP-9, serves as a useful statistically independent prognostic factor in ovarian cancer FIGO stage III, thus helping to identify ovarian cancer patients with an aggressive form of the disease.


Subject(s)
Matrix Metalloproteinase 9/biosynthesis , Ovarian Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Collagenases/metabolism , Enzyme Precursors/metabolism , Female , Gelatinases/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Metalloendopeptidases/metabolism , Middle Aged , Multivariate Analysis , Neoplasm Staging , Ovarian Neoplasms/pathology , Prognosis , Survival Rate
15.
Clin Cancer Res ; 7(8): 2396-404, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489818

ABSTRACT

Proteases are linked to the malignant phenotype of different solid tumors. Therefore, the expression of the matrix metalloproteinase (MMP)-2 and MMP-9 and of the serine protease urokinase-type plasminogen activator (uPA) and its inhibitor plasminogen activator inhibitor type 1 (PAI-1) in the progression of ovarian cancer was investigated. Gelatinolytic activity and protein expression of MMP-2 and MMP-9 were analyzed in tissue extracts of 19 cystadenomas and 18 low malignant potential (LMP) tumors, as well as 41 primary tumors of advanced ovarian cancer stage International Federation of Gynecology and Obstetrics IIIc/IV and their corresponding omentum metastases by quantitative gelatin zymography and Western blot. In the same tissue extracts, antigen levels of uPA and its inhibitor PAI-1 were determined by ELISA. Protein expression of pro-MMP-2 (72 kDa) and pro-MMP-9 (92 kDa as well as antigen levels of uPA and PAI-1 were low in benign ovarian tumors but increased significantly from LMP tumors to advanced ovarian cancers. The highest values of all of the proteolytic factors were detected in omentum metastases. Active MMP-2 enzyme (62 kDa) was detected only in ovarian cancer (66%) and corresponding metastases (93%) but never in benign or LMP tumors. The activation rate of MMP-2 to its active isoform was higher in the metastases. Comparing both proteolytic systems, higher PAI-1 concentrations were consistently found in cancers with high pro-MMP-9 expression. These data indicate that members of the plasminogen activator system, as well as the metalloproteinases MMP-2/9, increase with growing malignant potential of ovarian tumors. These findings are of particular relevance to the development of protease inhibitors as new therapeutic approaches in ovarian cancer.


Subject(s)
Endopeptidases/biosynthesis , Ovarian Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Blotting, Western , Disease Progression , Female , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/enzymology , Ovary/enzymology , Ovary/pathology , Plasminogen Activator Inhibitor 1/analysis , Statistics as Topic , Urokinase-Type Plasminogen Activator/biosynthesis
16.
Endocrinology ; 142(8): 3663-72, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459816

ABSTRACT

Gastric enterochromaffin-like cells produce histamine in response to the antral hormone gastrin and accumulate the biogenic amine in secretory organelles via vesicular monoamine transporter subtype 2. The putative effects of gastrin on vesicular monoamine transporter subtype 2 expression and promoter activity are poorly understood. In the present study we used highly enriched rat enterochromaffin-like cells (purity, >90%) and rat pheochromocytoma cells stably transfected with a gastrin/cholecystokinin B receptor to investigate the expression and transcriptional regulation of vesicular monoamine transporter subtype 2. Stimulation of vesicular monoamine transporter subtype 2 mRNA and protein expression was observed in isolated enterochromaffin-like cells after 3- to 7-h incubation with gastrin (10(-7) M), forskolin (10(-5) M), or ionomycin (10(-5) M). Deletion analysis of the rat vesicular monoamine transporter subtype 2 promoter defined the minimal promoter sequence necessary for full basal activity as a -121 bp segment upstream of exon 1 containing two Sp1 sites (-97 to -88 bp and -68 to -59 bp) and a cAMP-responsive element (-44 to -35 bp). Gastrin (10(-7) M) stimulated extracellular signal related kinase1/2 phosphorylation, activated Sp1 and cAMP-responsive element-binding protein, and further induced activity of the complete rat vesicular monoamine transporter subtype 2 promoter (-800 bp) in gastrin/cholecystokinin B receptor cells. The -121-bp fragment was able to confer full gastrin responsiveness, and site-directed mutagenesis of the Sp1 and cAMP-responsive element motifs demonstrated their crucial importance for basal and inducible activities. Comparison of promoter activity of histidine decarboxylase, chromogranin A, or vesicular monoamine transporter subtype 2 in transfected cell lines revealed significant differences in basal and gastrin-stimulated activities. Our current study provides the first evidence that gastrin directly stimulates the expression and promoter activity of vesicular monoamine transporter subtype 2. Sp1 and cAMP-responsive element-binding protein recognition motifs located within 121 bp upstream of exon 1 appear to be indispensable for full basal and inducible promoter activities. Diverging effects of gastrin on histidine decarboxylase, chromogranin A, and vesicular monoamine transporter subtype 2 promoter may account for the coordinated synthesis and storage of histamine in this neuroendocrine cell type.


Subject(s)
Gastrins/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Neuropeptides , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Animals , Chromogranin A , Chromogranins/genetics , Enterochromaffin Cells/drug effects , Enterochromaffin Cells/physiology , Histidine Decarboxylase/genetics , Immunohistochemistry , Membrane Glycoproteins/drug effects , PC12 Cells , RNA, Messenger/metabolism , Rats , Reference Values , Stimulation, Chemical , Transcription, Genetic/physiology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
18.
J Biol Chem ; 276(28): 26340-8, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11331280

ABSTRACT

The urokinase type plasminogen activator (uPA), together with its receptor uPAR and the plasminogen activator inhibitor type-1 (PAI-1) plays a pivotal role during tumor invasion and metastasis. Integrins, via interaction with the extracellular matrix (ECM), control cell adhesion and motility. The two systems are functionally linked because uPAR and PAI-1 bind to the ECM component vitronectin (VN). Because integrin signaling alters gene expression patterns, we investigated whether the expression levels of uPA, uPAR, and PAI-1 are affected by ECM/integrin interactions. Expression of uPA, uPAR, and PAI-1 was significantly enhanced when human ovarian cancer cells (OV-MZ-6) were cultivated on fibronectin or collagen type IV. In contrast, VN induced down-regulation of uPA and uPAR while increasing PAI-1 by up to 4-fold. VN-dependent decrease of uPA protein was paralleled by a significant reduction of uPA promoter activity that was even more pronounced upon alpha(v)beta(3) overexpression and depended on the presence of intact Rel protein-binding sites. The activity of Rel transcription factors was also significantly reduced upon alpha(v)beta(3)-mediated cell adhesion to VN. The activity of the Rel-unresponsive PAI-1 promoter was up to 5-fold induced as a function of alpha(v)beta(3)/VN interaction. Thus, the balance between available concentrations of uPA, uPAR, PAI-1, and integrins in human ovarian cancer cells might provide a switch within the regulation of their invasive phenotype.


Subject(s)
Ovarian Neoplasms/metabolism , Receptors, Vitronectin/metabolism , Urokinase-Type Plasminogen Activator/biosynthesis , Vitronectin/metabolism , Female , Humans , Tumor Cells, Cultured
20.
Mol Cell Biol ; 21(6): 2118-32, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238946

ABSTRACT

Migration of cells requires interactions with the extracellular matrix mediated, in part, by integrins, proteases, and their receptors. Previous studies have shown that beta(3)-integrin interacts with the urokinase-type plasminogen activator receptor (u-PAR) at the cell surface. Since integrins mediate signaling into the cell, the current study was undertaken to determine if in addition beta(3)-integrin regulates u-PAR expression. Overexpression of beta(3)-integrin in CHO cells, which are avid expressers of the receptor, downregulated u-PAR protein and mRNA expression. The u-PAR promoter (-1,469 bp) that is normally constitutively active in CHO cells was downregulated by induced beta(3)-integrin expression. A region between -398 and -197 bp of the u-PAR promoter was critical for beta(3)-integrin-induced downregulation of u-PAR promoter activity. Deletion of the PEA3/ets motif at -248 bp substantially impaired the ability of beta(3)-integrin to downregulate the u-PAR promoter, suggesting that the PEA3/ets site acts as a silencing element. An expression vector encoding the transcription factor PEA3 caused inhibition of the wild-type but not the PEA3/ets-deleted u-PAR promoter. The PEA3/ets site bound nuclear factors from CHO cells specifically, but binding was enhanced when beta(3)-integrin was overexpressed. A PEA3 antibody inhibited DNA-protein complex formation, indicating the presence of PEA3. Downregulation of the u-PAR promoter was achieved by the beta(3)A-integrin isoform but not by other beta(3)-integrin isoforms and required the cytoplasmic membrane NITY(759) motif. Moreover, overexpression of the short but not the long isoform of the beta(3)-integrin adapter protein beta(3)-endonexin blocked u-PAR promoter activity through the PEA3/ets binding site. Thus, besides the physical interaction of beta(3)-integrin and u-PAR at the cell surface, beta(3) signaling is implicated in the regulation of u-PAR gene transcription, suggesting a mutual regulation of adhesion and proteolysis receptors.


Subject(s)
Antigens, CD/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, Cell Surface/genetics , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Amino Acid Sequence , Animals , Antigens, CD/genetics , Base Sequence , Binding Sites , CHO Cells , Cricetinae , Cytoplasm/metabolism , Down-Regulation , Gene Expression Regulation , Integrin beta3 , Molecular Sequence Data , Nuclear Proteins , Platelet Membrane Glycoproteins/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...