Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Eng Part A ; 26(13-14): 747-758, 2020 07.
Article in English | MEDLINE | ID: mdl-32598229

ABSTRACT

It is well known that during ovarian cancer progression, the omentum transforms from a thin lacy organ to a thick tougher tissue. However, the mechanisms regulating this transformation and the implications of the altered microenvironment on ovarian cancer progression remain unclear. To address these questions, the global and local concentrations of collagen I were determined for normal and metastatic human omentum. Collagen I was increased 5.3-fold in omenta from ovarian cancer patients and localized to areas of activated fibroblasts rather than regions with a high density of cancer cells. Transforming growth factor beta 1 (TGFß1) was detected in ascites from ovarian cancer patients (4 ng/mL), suggesting a potential role for TGFß1 in the observed increase in collagen. Treatment with TGFß1 induced fibroblast activation, proliferation, and collagen deposition in mouse omental explants and an in vitro model with human omental fibroblasts. Finally, the impact of increased collagen I on ovarian cancer cells was determined by examining proliferation on collagen I gels formulated to mimic normal and cancerous omenta. While collagen density alone had no impact on proliferation, a synergistic effect was observed with collagen density and heparin-binding epidermal growth factor treatment. These results suggest that TGFß1 induces collagen deposition from the resident fibroblasts in the omentum and that this altered microenvironment impacts cancer cell response to growth factors found in ascites. Impact statement Using quantitative analysis of patient samples, in vitro models of the metastatic ovarian cancer microenvironment were designed with pathologically relevant collagen densities and growth factor concentrations. Studies in these models support a mechanism where transforming growth factor ß1 in the ascites fluid induces omental fibroblast proliferation, activation, and deposition of collagen I, which then impacts tumor cell proliferation in response to additional ascites growth factors such as heparin-binding epidermal growth factor. This approach can be used to dissect mechanisms involved in microenvironmental modeling in multiple disease applications.


Subject(s)
Collagen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Humans , In Situ Hybridization , Ovarian Neoplasms/metabolism , Transforming Growth Factor beta1/pharmacology
2.
Cancer Lett ; 458: 92-101, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31129149

ABSTRACT

High-grade serous ovarian cancer (HGSOC) metastasizes when tumor spheroids detach from the primary tumor and re-attach throughout the peritoneal cavity. Once the cancer cells have implanted in these new sites, the development of metastatic lesions is dependent on the disaggregation of cancer cells from the spheroids and subsequent expansion across the collagenous extracellular matrix (ECM). As HGSOC progresses an increase in alternatively activated macrophages (AAMs) in the surrounding ascites fluid has been observed and AAMs have been shown to enhance tumor invasion and growth in a wide range of cancers. We hypothesized that soluble factors from AAMs in the peritoneal microenvironment promote the disaggregation of ovarian cancer spheroids across the underlying ECM. We determined that co-culture with AAMs significantly increased HGSOC spheroid spreading across a collagen matrix. Multivariate modeling identified AAM-derived factors that correlated with enhanced spread of HGSOC spheroids and experimental validation showed that each individual cell line responded to a distinct AAM-derived factor (FLT3L, leptin, or HB-EGF). Despite this ligand-level heterogeneity, we determined that the AAM-derived factors utilized a common signaling pathway to induce spheroid spreading: JAK2/STAT3 activation followed by MMP-9 mediated spreading. Furthermore, immunostaining demonstrated that FLT3, LEPR, EGFR, and pSTAT3 were upregulated in metastases in HGSOC patients, with substantial patient-to-patient heterogeneity. These results suggest that inhibiting individual soluble factors will not inhibit AAM-induced effects across a broad group of patients; instead, the downstream JAK2/STAT3/MMP-9 pathway should be examined as potential therapeutic targets to slow metastasis in ovarian cancer.


Subject(s)
Janus Kinase 2/metabolism , Macrophages/pathology , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Adolescent , Adult , Cell Line, Tumor , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Female , Humans , Janus Kinase 2/immunology , Macrophage Activation , Macrophages/immunology , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasm Metastasis , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Receptors, Leptin/immunology , Receptors, Leptin/metabolism , STAT3 Transcription Factor/immunology , Signal Transduction , Spheroids, Cellular , Up-Regulation , Young Adult , fms-Like Tyrosine Kinase 3/immunology , fms-Like Tyrosine Kinase 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...