Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 10877, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616784

ABSTRACT

Epithelial ovarian cancer (EOC) metastasis occurs by exfoliation of cells and multicellular aggregates (MCAs) from the tumor into the peritoneal cavity, adhesion to and retraction of peritoneal mesothelial cells and subsequent anchoring. Elevated levels of lysophosphatidic acid (LPA) have been linked to aberrant cell proliferation, oncogenesis, and metastasis. LPA disrupts junctional integrity and epithelial cohesion in vitro however, the fate of free-floating cells/MCAs and the response of host peritoneal tissues to LPA remain unclear. EOC MCAs displayed significant LPA-induced changes in surface ultrastructure with the loss of cell surface protrusions and poor aggregation, resulting in increased dissemination of small clusters compared to untreated control MCAs. LPA also diminished the adhesive capacity of EOC single cells and MCAs to murine peritoneal explants and impaired MCA survival and mesothelial clearance competence. Peritoneal tissues from healthy mice injected with LPA exhibited enhanced mesothelial surface microvilli. Ultrastructural alterations were associated with restricted peritoneal susceptibility to metastatic colonization by single cells as well as epithelial-type MCAs. The functional consequence is an LPA-induced dissemination of small mesenchymal-type clusters, promoting a miliary mode of peritoneal seeding that complicates surgical removal and is associated with worse prognosis.


Subject(s)
Carcinoma, Ovarian Epithelial/pathology , Cell Aggregation/drug effects , Epithelial Cells/drug effects , Lysophospholipids/pharmacology , Ovarian Neoplasms/pathology , Animals , Carcinoma, Ovarian Epithelial/secondary , Cell Adhesion/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microvilli/drug effects , Peritoneal Neoplasms/secondary , Tumor Microenvironment
2.
Neoplasia ; 20(6): 621-631, 2018 06.
Article in English | MEDLINE | ID: mdl-29754071

ABSTRACT

Ovarian cancer, the most deadly gynecological malignancy in U.S. women, metastasizes uniquely, spreading through the peritoneal cavity and often generating widespread metastatic sites before diagnosis. The vast majority of ovarian cancer cases occur in women over 40 and the median age at diagnosis is 63. Additionally, elderly women receive poorer prognoses when diagnosed with ovarian cancer. Despite age being a significant risk factor for the development of this cancer, there are little published data which address the impact of aging on ovarian cancer metastasis. Here we report that the aged host is more susceptible to metastatic success using two murine syngeneic allograft models of ovarian cancer metastasis. This age-related increase in metastatic tumor burden corresponds with an increase in tumor infiltrating lymphocytes (TILs) in tumor-bearing mice and alteration of B cell-related pathways in gonadal adipose tissue. Based on this work, further studies elucidating the status of B cell TILs in mouse models of metastasis and human tumors in the context of aging are warranted.


Subject(s)
Adipose Tissue/pathology , Aging/pathology , Allografts/pathology , Neoplasm Metastasis/pathology , Ovarian Neoplasms/pathology , Adult , Aged , Animals , Cell Line , Female , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Prognosis , Risk Factors , Tumor Burden/physiology , Young Adult
3.
Methods Cell Biol ; 143: 79-95, 2018.
Article in English | MEDLINE | ID: mdl-29310793

ABSTRACT

This chapter highlights methods for visualization and analysis of extracellular matrix (ECM) proteins, with particular emphasis on collagen type I, the most abundant protein in mammals. Protocols described range from advanced imaging of complex in vivo matrices to simple biochemical analysis of individual ECM proteins. The first section of this chapter describes common methods to image ECM components and includes protocols for second harmonic generation, scanning electron microscopy, and several histological methods of ECM localization and degradation analysis, including immunohistochemistry, Trichrome staining, and in situ zymography. The second section of this chapter details both a common transwell invasion assay and a novel live imaging method to investigate cellular behavior with respect to collagen and other ECM proteins of interest. The final section consists of common electrophoresis-based biochemical methods that are used in analysis of ECM proteins. Use of the methods described herein will enable researchers to gain a greater understanding of the role of ECM structure and degradation in development and matrix-related diseases such as cancer and connective tissue disorders.


Subject(s)
Collagen Type I/ultrastructure , Extracellular Matrix/ultrastructure , Molecular Imaging/methods , Proteolysis , Staining and Labeling/methods , Animals , Collagen Type I/chemistry , Connective Tissue Diseases/etiology , Connective Tissue Diseases/pathology , Extracellular Matrix/chemistry , Humans , Immunohistochemistry/methods , Microscopy, Electron, Scanning/instrumentation , Microscopy, Electron, Scanning/methods , Molecular Imaging/instrumentation , Staining and Labeling/instrumentation
4.
Anal Bioanal Chem ; 410(5): 1583-1594, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29282499

ABSTRACT

Cancer metastasis risk increases in older individuals, but the mechanisms for this risk increase are unclear. Many peritoneal cancers, including ovarian cancer, preferentially metastasize to peritoneal fat depots. However, there is a dearth of studies exploring aged peritoneal adipose tissue in the context of cancer. Because adipose tissue produces signals which influence several diseases including cancer, proteomics of adipose tissue in aged and young mice may provide insight into metastatic mechanisms. We analyzed mesenteric, omental, and uterine adipose tissue groups from the peritoneal cavities of young and aged C57BL/6J mouse cohorts with a low-fraction SDS-PAGE gelLC-MS/MS method. We identified 2308 protein groups and quantified 2167 groups, among which several protein groups showed twofold or greater abundance differences between the aged and young cohorts. Cancer-related gene products previously identified as significant in another age-related study were found altered in this study. Several gene products known to suppress proliferation and cellular invasion were found downregulated in the aged cohort, including R-Ras, Arid1a, and heat shock protein ß1. In addition, multiple protein groups were identified within single cohorts, including the proteins Cd11a, Stat3, and Ptk2b. These data suggest that adipose tissue is a strong candidate for analysis to identify possible contributors to cancer metastasis in older subjects. The results of this study, the first of its kind using uterine adipose tissue, contribute to the understanding of the role of adipose tissue in age-related alteration of oncogenic pathways, which may help elucidate the mechanisms of increased metastatic tumor burden in the aged. Graphical abstract We analyzed mesenteric, omental, and uterine adipose tissue groups from the peritoneal cavities of young and aged C57BL/6J mouse cohorts with a low-fraction SDS-PAGE gelLC-MS/MS method. These fat depots are preferential sites for many peritoneal cancers. The results of this study, the first of its kind using uterine adipose tissue, contribute to the understanding of the role of adipose tissue in age-related alteration of oncogenic pathways, which may help elucidate the mechanisms of increased metastatic tumor burden in the aged.


Subject(s)
Adipose Tissue, White/chemistry , Ovarian Neoplasms , Proteomics , Adipose Tissue, White/pathology , Age Factors , Animals , Electrophoresis, Polyacrylamide Gel , Female , Humans , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/physiopathology , Ovarian Neoplasms/secondary
5.
Cancer Lett ; 411: 74-81, 2017 12 28.
Article in English | MEDLINE | ID: mdl-28964786

ABSTRACT

Ovarian cancer is the fifth leading cause of cancer deaths in U.S. women and the deadliest gynecologic malignancy. This lethality is largely due to the fact that most cases are diagnosed at metastatic stages of the disease when the prognosis is poor. Epidemiologic studies consistently demonstrate that parous women have a reduced risk of developing ovarian cancer, with a greater number of births affording greater protection; however little is known about the impact of parity on ovarian cancer metastasis. Here we report that multiparous mice are less susceptible to ovarian cancer metastasis in an age-matched syngeneic murine allograft model. Interferon pathways were found to be upregulated in healthy adipose tissue of multiparous mice, suggesting a possible mechanism for the multiparous-related protective effect against metastasis. This protective effect was found to be lost with age. Based on this work, future studies exploring therapeutic strategies which harness the multiparity-associated protective effect demonstrated here are warranted.


Subject(s)
Adipose Tissue/metabolism , Interferons/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Parity , Peritoneum/metabolism , Adipose Tissue/pathology , Allografts , Animals , Carcinoma, Ovarian Epithelial , Disease Models, Animal , Disease Susceptibility , Female , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Peritoneum/pathology , Pregnancy , Risk Factors
6.
Neoplasia ; 19(7): 549-563, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28601643

ABSTRACT

Epithelial ovarian carcinoma spreads via shedding of cells and multicellular aggregates (MCAs) from the primary tumor into peritoneal cavity, with subsequent intraperitoneal tumor cell:mesothelial cell adhesion as a key early event in metastatic seeding. Evaluation of human tumor extracts and tissues confirms that well-differentiated ovarian tumors express abundant E-cadherin (Ecad), whereas advanced lesions exhibit upregulated N-cadherin (Ncad). Two expression patterns are observed: "mixed cadherin," in which distinct cells within the same tumor express either E- or Ncad, and "hybrid cadherin," wherein single tumor cell(s) simultaneously expresses both cadherins. We demonstrate striking cadherin-dependent differences in cell-cell interactions, MCA formation, and aggregate ultrastructure. Mesenchymal-type Ncad+ cells formed stable, highly cohesive solid spheroids, whereas Ecad+ epithelial-type cells generated loosely adhesive cell clusters covered by uniform microvilli. Generation of "mixed cadherin" MCAs using fluorescently tagged cell populations revealed preferential sorting into cadherin-dependent clusters, whereas mixing of cell lines with common cadherin profiles generated homogeneous aggregates. Recapitulation of the "hybrid cadherin" Ecad+/Ncad+ phenotype, via insertion of the CDH2 gene into Ecad+ cells, resulted in the ability to form heterogeneous clusters with Ncad+ cells, significantly enhanced adhesion to organotypic mesomimetic cultures and peritoneal explants, and increased both migration and matrix invasion. Alternatively, insertion of CDH1 gene into Ncad+ cells greatly reduced cell-to-collagen, cell-to-mesothelium, and cell-to-peritoneum adhesion. Acquisition of the hybrid cadherin phenotype resulted in altered MCA surface morphology with increased surface projections and increased cell proliferation. Overall, these findings support the hypothesis that MCA cadherin composition impacts intraperitoneal cell and MCA dynamics and thereby affects ultimate metastatic success.


Subject(s)
Cadherins/genetics , Gene Expression , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cadherins/metabolism , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Neoplasm Staging , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/ultrastructure , Phenotype , Tissue Array Analysis
7.
Cancer Res ; 75(23): 5046-57, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26573796

ABSTRACT

Epithelial ovarian cancer (EOC) is the leading cause of death from gynecologic malignancy, with high mortality attributable to widespread intraperitoneal metastases. Recent meta-analyses report an association between obesity, ovarian cancer incidence, and ovarian cancer survival, but the effect of obesity on metastasis has not been evaluated. The objective of this study was to use an integrative approach combining in vitro, ex vivo, and in vivo studies to test the hypothesis that obesity contributes to ovarian cancer metastatic success. Initial in vitro studies using three-dimensional mesomimetic cultures showed enhanced cell-cell adhesion to the lipid-loaded mesothelium. Furthermore, in an ex vivo colonization assay, ovarian cancer cells exhibited increased adhesion to mesothelial explants excised from mice modeling diet-induced obesity (DIO), in which they were fed a "Western" diet. Examination of mesothelial ultrastructure revealed a substantial increase in the density of microvilli in DIO mice. Moreover, enhanced intraperitoneal tumor burden was observed in overweight or obese animals in three distinct in vivo models. Further histologic analyses suggested that alterations in lipid regulatory factors, enhanced vascularity, and decreased M1/M2 macrophage ratios may account for the enhanced tumorigenicity. Together, these findings show that obesity potently affects ovarian cancer metastatic success, which likely contributes to the negative correlation between obesity and ovarian cancer survival.


Subject(s)
Macrophages/pathology , Obesity/pathology , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Lipogenesis , Macrophages/immunology , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Obesity/immunology , Obesity/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism
8.
Gynecol Oncol ; 138(2): 372-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050922

ABSTRACT

OBJECTIVE: Genomic studies of ovarian cancer (OC) cell lines frequently used in research revealed that these cells do not fully represent high-grade serous ovarian cancer (HGSOC), the most common OC histologic type. However, OC lines that appear to genomically resemble HGSOC have not been extensively used and their growth characteristics in murine xenografts are essentially unknown. METHODS: To better understand growth patterns and characteristics of HGSOC cell lines in vivo, CAOV3, COV362, KURAMOCHI, NIH-OVCAR3, OVCAR4, OVCAR5, OVCAR8, OVSAHO, OVKATE, SNU119 and UWB1.289 cells were assessed for tumor formation in nude mice. Cells were injected intraperitoneally (i.p.) or subcutaneously (s.c.) in female athymic nude mice and allowed to grow (maximum of 90 days) and tumor formation was analyzed. All tumors were sectioned and assessed using H&E staining and immunohistochemistry for p53, PAX8 and WT1 expression. RESULTS: Six lines (OVCAR3, OVCAR4, OVCAR5, OVCAR8, CAOV3, and OVSAHO) formed i.p xenografts with HGSOC histology. OVKATE and COV362 formed s.c. tumors only. Rapid tumor formation was observed for OVCAR3, OVCAR5 and OVCAR8, but only OVCAR8 reliably formed ascites. Tumors derived from OVCAR3, OVCAR4, and OVKATE displayed papillary features. Of the 11 lines examined, three (Kuramochi, SNU119 and UWB1.289) were non-tumorigenic. CONCLUSIONS: Our findings help further define which HGSOC cell models reliably generate tumors and/or ascites, critical information for preclinical drug development, validating in vitro findings, imaging and prevention studies by the OC research community.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Disease Models, Animal , Ovarian Neoplasms/pathology , Animals , Cell Growth Processes , Cell Line, Tumor , Cystadenocarcinoma, Serous/metabolism , Female , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Grading , Ovarian Neoplasms/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , WT1 Proteins/biosynthesis
9.
Breast Cancer Res ; 15(5): R91, 2013.
Article in English | MEDLINE | ID: mdl-24074261

ABSTRACT

INTRODUCTION: The Rho GTPase Cdc42 is overexpressed and hyperactivated in breast tumors compared to normal breast tissue. Cdc42 regulates key processes that are critical for mammary gland morphogenesis and become disrupted during the development, progression, and metastasis of breast cancer. However, the contribution of Cdc42 to normal and neoplastic mammary gland development in vivo remains poorly understood. We were therefore interested in investigating the effects of Cdc42 overexpression on mammary gland morphogenesis as a first step toward understanding how its overexpression may contribute to mammary tumorigenesis. METHODS: We developed a tetracycline-regulatable Cdc42 overexpression mouse model in which Cdc42 can be inducibly overexpressed in the developing mammary gland. The effects of Cdc42 overexpression during postnatal mammary gland development were investigated using in vivo and in vitro approaches, including morphometric analysis of wholemounted mammary glands, quantification of histological markers, and primary mammary epithelial cell (MEC) functional and biochemical assays. RESULTS: Analysis of Cdc42-overexpressing mammary glands revealed abnormal terminal end bud (TEB) morphologies, characterized by hyperbudding and trifurcation, and increased side branching within the ductal tree. Quantification of markers of proliferation and apoptosis suggested that these phenotypes were not due to increased cell proliferation or survival. Rather, Cdc42 overexpressing MECs were more migratory and contractile and formed dysmorphic, invasive acini in three-dimensional cultures. Cdc42 and RhoA activities, phosphorylated myosin light chain, and MAPK signaling, which contribute to migration and invasion, were markedly elevated in Cdc42 overexpressing MECs. Interestingly, Cdc42 overexpressing mammary glands displayed several features associated with altered epithelial-stromal interactions, which are known to regulate branching morphogenesis. These included increased stromal thickness and collagen deposition, and stromal cells isolated from Cdc42 overexpressing mammary glands exhibited elevated mRNA expression of extracellular matrix proteins and remodeling enzymes. CONCLUSIONS: These data suggest that Cdc42 overexpression disrupts mammary gland branching morphogenesis by altering Rho GTPase and MAPK signaling, leading to increased MEC contractility and migration in association with stromal alterations. Our studies provide insight into how aberrant Cdc42 expression may contribute to mammary tumorigenesis.


Subject(s)
Gene Expression , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , cdc42 GTP-Binding Protein/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , MAP Kinase Signaling System , Mice , Mice, Transgenic , Stromal Cells/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...