Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 3546, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669559

ABSTRACT

Advanced ovarian cancer usually spreads to the omentum. However, the omental cell-derived molecular determinants modulating its progression have not been thoroughly characterized. Here, we show that circulating ITLN1 has prognostic significance in patients with advanced ovarian cancer. Further studies demonstrate that ITLN1 suppresses lactotransferrin's effect on ovarian cancer cell invasion potential and proliferation by decreasing MMP1 expression and inducing a metabolic shift in metastatic ovarian cancer cells. Additionally, ovarian cancer-bearing mice treated with ITLN1 demonstrate marked decrease in tumor growth rates. These data suggest that downregulation of mesothelial cell-derived ITLN1 in the omental tumor microenvironment facilitates ovarian cancer progression.


Subject(s)
Carcinoma, Ovarian Epithelial/secondary , Cytokines/metabolism , Lectins/metabolism , Omentum/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Animals , Carcinoma, Ovarian Epithelial/blood , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor/transplantation , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cytokines/administration & dosage , Cytokines/blood , Disease Models, Animal , Down-Regulation , Female , GPI-Linked Proteins/administration & dosage , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lactoferrin/metabolism , Lectins/administration & dosage , Lectins/blood , Matrix Metalloproteinase 1/metabolism , Mice , Neoplasm Invasiveness/pathology , Ovarian Neoplasms/blood , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Ovary , Recombinant Proteins/administration & dosage , Survival Rate , Tumor Microenvironment
3.
Cancers (Basel) ; 12(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906456

ABSTRACT

Uterine serous carcinoma (USC) is the most aggressive form of endometrial cancer, with poor survival rates and high recurrence risk. Therefore, the purpose of this study was to identify therapeutic targets that could aid in the management of USC. By analyzing endometrial cancer samples from The Cancer Genome Atlas (TCGA), we found Ubiquitin Carboxyl-Terminal Hydrolase L1 (UCHL1) to be highly expressed in USC and to correlate with poorer overall survival. UCHL1 silencing reduced cell proliferation in vitro and in vivo, cyclin B1 protein levels and cell cycle progression. Further studies showed that UCHL1 interacts with cyclin B1 and increases cyclin B1 protein stability by deubiquitination. Treatment of USC-bearing mice with the UCHL1-specific inhibitor reduced tumor growth and improved overall survival. Our findings suggest that cyclin B1 is a novel target of UCHL1 and targeting UCHL1 is a potential therapeutic strategy for USC.

4.
Neoplasia ; 21(11): 1073-1084, 2019 11.
Article in English | MEDLINE | ID: mdl-31734628

ABSTRACT

The role of prostaglandin (PG) F2α has been scarcely studied in cancer. We have identified a new function for PGF2α in ovarian cancer, stimulating the production of Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1). We show that this induction increases cell plasticity and proliferation, enhancing tumor growth through PMEPA1. Thus, PMEPA1 overexpression in ovarian carcinoma cells, significantly increased cell proliferation rates, whereas PMEPA1 silencing decreased proliferation. In addition, PMEPA1 overexpression buffered TGFß signaling, via reduction of SMAD-dependent signaling. PMEPA1 overexpressing cells acquired an epithelial morphology, associated with higher E-cadherin expression levels while ß-catenin nuclear translocation was inhibited. Notwithstanding, high PMEPA1 levels also correlated with epithelial to mesenchymal transition markers, such as vimentin and ZEB1, allowing the cells to take advantage of both epithelial and mesenchymal characteristics, gaining in cell plasticity and adaptability. Interestingly, in mouse xenografts, PMEPA1 overexpressing ovarian cells had a clear survival and proliferative advantage, resulting in higher metastatic capacity, while PMEPA1 silencing had the opposite effect. Furthermore, high PMEPA1 expression in a cohort of advanced ovarian cancer patients was observed, correlating with E-cadherin expression. Most importantly, high PMEPA1 mRNA levels were associated with lower patient survival.

5.
Nat Commun ; 10(1): 4369, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554806

ABSTRACT

Chromatin accessibility data can elucidate the developmental origin of cancer cells and reveal the enhancer landscape of key oncogenic transcriptional regulators. We develop a computational strategy called PSIONIC (patient-specific inference of networks informed by chromatin) to combine chromatin accessibility data with large tumor expression data and model the effect of enhancers on transcriptional programs in multiple cancers. We generate a new ATAC-seq data profiling chromatin accessibility in gynecologic and basal breast cancer cell lines and apply PSIONIC to 723 patient and 96 cell line RNA-seq profiles from ovarian, uterine, and basal breast cancers. Our computational framework enables us to share information across tumors to learn patient-specific TF activities, revealing regulatory differences between and within tumor types. PSIONIC-predicted activity for MTF1 in cell line models correlates with sensitivity to MTF1 inhibition, showing the potential of our approach for personalized therapy. Many identified TFs are significantly associated with survival outcome. To validate PSIONIC-derived prognostic TFs, we perform immunohistochemical analyses in 31 uterine serous tumors for ETV6 and 45 basal breast tumors for MITF and confirm that the corresponding protein expression patterns are also significantly associated with prognosis.


Subject(s)
Breast Neoplasms/genetics , Chromatin/genetics , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Humans , Kaplan-Meier Estimate , Transcription Factors/genetics , Transcription Factor MTF-1
6.
Nat Commun ; 8: 15509, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28593951

ABSTRACT

Matrix metalloproteinases (MMPs) contribute to the breakdown of tissue structures such as the basement membrane, promoting tissue fibrosis. Here we developed an electrospun membrane biofunctionalized with a fragment of the laminin ß1-chain to modulate the expression of MMP2 in this context. We demonstrate that interfacing of the ß1-fragment with the mesothelium of the peritoneal membrane via a biomaterial abrogates the release of active MMP2 in response to transforming growth factor ß1 and rescues tissue integrity ex vivo and in vivo in a mouse model of peritoneal fibrosis. Importantly, our data demonstrate that the membrane inhibits MMP2 expression. Changes in the expression of epithelial-to-mesenchymal transition (EMT)-related molecules further point towards a contribution of the modulation of EMT. Biomaterial-based presentation of regulatory basement membrane signals directly addresses limitations of current therapeutic approaches by enabling a localized and specific method to counteract MMP2 release applicable to a broad range of therapeutic targets.


Subject(s)
Biocompatible Materials/chemistry , Extracellular Matrix/metabolism , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Animals , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelium/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Integrin alpha3beta1/metabolism , Laminin/metabolism , Mammary Glands, Human/cytology , Matrix Metalloproteinase 2/metabolism , Membranes, Artificial , Mice , Peritoneum/metabolism , Protein Binding , Signal Transduction
7.
J Pathol ; 242(2): 140-151, 2017 06.
Article in English | MEDLINE | ID: mdl-28247413

ABSTRACT

Peritoneal dissemination is the primary metastatic route of ovarian cancer (OvCa), and is often accompanied by the accumulation of ascitic fluid. The peritoneal cavity is lined by mesothelial cells (MCs), which can be converted into carcinoma-associated fibroblasts (CAFs) through mesothelial-to-mesenchymal transition (MMT). Here, we demonstrate that MCs isolated from ascitic fluid (AFMCs) of OvCa patients with peritoneal implants also undergo MMT and promote subcutaneous tumour growth in mice. RNA sequencing of AFMCs revealed that MMT-related pathways - including transforming growth factor (TGF)-ß signalling - are differentially regulated, and a gene signature was verified in peritoneal implants from OvCa patients. In a mouse model, pre-induction of MMT resulted in increased peritoneal tumour growth, whereas interfering with the TGF-ß receptor reduced metastasis. MC-derived CAFs showed activation of Smad-dependent TGF-ß signalling, which was disrupted in OvCa cells, despite their elevated TGF-ß production. Accordingly, targeting Smad-dependent signalling in the peritoneal pre-metastatic niche in mice reduced tumour colonization, suggesting that Smad-dependent MMT could be crucial in peritoneal carcinomatosis. Together, these results indicate that bidirectional communication between OvCa cells and MC-derived CAFs, via TGF-ß-mediated MMT, seems to be crucial to form a suitable metastatic niche. We suggest MMT as a possible target for therapeutic intervention and a potential source of biomarkers for improving OvCa diagnosis and/or prognosis. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma/secondary , Epithelial-Mesenchymal Transition , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Animals , Ascites/pathology , Ascitic Fluid/pathology , Carcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Epithelial Cells/pathology , Female , Fibroblasts/pathology , Humans , Mice , Ovarian Neoplasms/complications , Peritoneal Neoplasms/pathology , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Sequence Analysis, RNA , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
8.
J Pathol ; 239(1): 48-59, 2016 May.
Article in English | MEDLINE | ID: mdl-27071481

ABSTRACT

Peritoneal adhesions (PAs) are fibrotic bands formed between bowel loops, solid organs, and the parietal peritoneum, which may appear following surgery, infection or endometriosis. They represent an important health problem with no effective treatment. Mesothelial cells (MCs) line the peritoneal cavity and undergo a mesothelial-to-mesenchymal transition (MMT) under pathological conditions, transforming into myofibroblasts, which are abundant in peritoneal fibrotic tissue. The aim of this study was to investigate if peritoneal MCs undergo a MMT contributing to the formation of post-surgical adhesions. Biopsies from patients with PAs were analysed by immunohistochemistry, immunofluorescence, and quantitative RT-PCR. A mouse model of PAs based on ischaemic buttons was used to modulate MMT by blocking the transforming growth factor-beta (TGF-ß) pathway. The severity of adhesions and MMT-related marker expression were studied. We observed myofibroblasts derived from the conversion of MCs in submesothelial areas of patients with PAs. In addition, MMT-related markers were dysregulated in adhesion zones when compared to distant normal peritoneal tissue of the same patient. In animal experiments, blockage of TGF-ß resulted in molecular reprogramming of markers related to the mesenchymal conversion of MCs and in a significant decrease in the severity of the adhesions. These data indicate for the first time that MMT is involved in PA pathogenesis. This finding opens new therapeutic strategies to interfere with adhesion formation by modulating MMT with a wide range of pharmacological agents.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Tissue Adhesions/etiology , Actins/metabolism , Adult , Aged , Animals , Calbindin 2/metabolism , Female , Fibroblasts/physiology , Humans , Keratins/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Peptide Fragments/pharmacology , Peritoneum , Receptors, Transforming Growth Factor beta , Smad3 Protein/metabolism , Tissue Adhesions/pathology , Young Adult
9.
Cancers (Basel) ; 7(4): 1994-2011, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26426054

ABSTRACT

Solid tumors are complex and unstructured organs that, in addition to cancer cells, also contain other cell types. Carcinoma-associated fibroblasts (CAFs) represent an important population in the tumor microenviroment and participate in several stages of tumor progression, including cancer cell migration/invasion and metastasis. During peritoneal metastasis, cancer cells detach from the primary tumor, such as ovarian or gastrointestinal, disseminate through the peritoneal fluid and colonize the peritoneum. Tumor cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity, then colonizing the submesothelial compact zone where CAFs accumulate. CAFs may derive from different sources depending on the surrounding metastatic niche. In peritoneal metastasis, a sizeable subpopulation of CAFs originates from MCs through a mesothelial-to-mesenchymal transition (MMT), which promotes adhesion, invasion, vascularization and subsequent tumor growth. The bidirectional communication between cancer cells and MC-derived CAFs via secretion of a wide range of cytokines, growth factors and extracellular matrix components seems to be crucial for the establishment and progression of the metastasis in the peritoneum. This manuscript provides a comprehensive review of novel advances in understanding how peritoneal CAFs provide cancer cells with a supportive microenvironment, as well as the development of future therapeutic approaches by interfering with the MMT in the peritoneum.

10.
J Pathol ; 231(4): 517-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24114721

ABSTRACT

Peritoneal dissemination is a frequent metastatic route for cancers of the ovary and gastrointestinal tract. Tumour cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity. Metastases are influenced by carcinoma-associated fibroblasts (CAFs), a cell population that derives from different sources. Hence, we investigated whether MCs, through mesothelial-mesenchymal transition (MMT), were a source of CAFs during peritoneal carcinomatosis and whether MMT affected the adhesion and invasion of tumour cells. Biopsies from patients with peritoneal dissemination revealed the presence of myofibroblasts expressing mesothelial markers in the proximity of carcinoma implants. Prominent new vessel formation was observed in the peritoneal areas harbouring tumour cells when compared with tumour-free regions. The use of a mouse model of peritoneal dissemination confirmed the myofibroblast conversion of MCs and the increase in angiogenesis at places of tumour implants. Treatment of omentum MCs with conditioned media from carcinoma cell cultures resulted in phenotype changes reminiscent of MMT. Adhesion experiments demonstrated that MMT enhanced the binding of cancer cells to MCs in a ß1-integrin-dependent manner. Scanning electron microscopy imaging showed that the enhanced adhesion was mostly due to increased cell-cell interaction and not to a mere matrix exposure. Invasion assays suggested a reciprocal stimulation of the invasive capacity of tumour cells and MCs. Our results demonstrate that CAFs can derive from mesothelial cells during peritoneal metastasis. We suggest that MMT renders the peritoneum more receptive for tumour cell attachment/invasion and contributes to secondary tumour growth by promoting its vascularization.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Fibroblasts/pathology , Peritoneal Neoplasms/secondary , Animals , Biopsy , Cell Adhesion , Cell Line, Tumor , Colorectal Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Epithelial Cells/pathology , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Epithelial-Mesenchymal Transition/drug effects , Female , Fibroblasts/physiology , Heterografts , Humans , Mice , Mice, Nude , Microscopy, Electron, Scanning , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/blood supply , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/ultrastructure
11.
PLoS One ; 8(4): e60776, 2013.
Article in English | MEDLINE | ID: mdl-23585849

ABSTRACT

Vascular endothelial growth factor (VEGF) is up-regulated during mesothelial to mesenchymal transition (MMT) and has been associated with peritoneal membrane dysfunction in peritoneal dialysis (PD) patients. It has been shown that normal and malignant mesothelial cells (MCs) express VEGF receptors (VEGFRs) and co-receptors and that VEGF is an autocrine growth factor for mesothelioma. Hence, we evaluated the expression patterns and the functional relevance of the VEGF/VEGFRs/co-receptors axis during the mesenchymal conversion of MCs induced by peritoneal dialysis. Omentum-derived MCs treated with TGF-ß1 plus IL-1ß (in vitro MMT) and PD effluent-derived MCs with non-epithelioid phenotype (ex vivo MMT) showed down-regulated expression of the two main receptors Flt-1/VEGFR-1 and KDR/VEGFR-2, whereas the co-receptor neuropilin-1 (Nrp-1) was up-regulated. The expression of the Nrp-1 ligand semaphorin-3A (Sema-3A), a functional VEGF competitor, was repressed throughout the MMT process. These expression pattern changes were accompanied by a reduction of the proliferation capacity and by a parallel induction of the invasive capacity of MCs that had undergone an in vitro or ex vivo MMT. Treatment with neutralizing anti-VEGF or anti-Nrp-1 antibodies showed that these molecules played a relevant role in cellular proliferation only in naïve omentum-derived MCs. Conversely, treatment with these blocking antibodies, as well as with recombinant Sema-3A, indicated that the switched VEGF/VEGFRs/co-receptors axis drove the enhanced invasion capacity of MCs undergoing MMT. In conclusion, the expression patterns of VEGFRs and co-receptors change in MCs during MMT, which in turn would determine their behaviour in terms of proliferation and invasion in response to VEGF.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Neuropilin-1/genetics , Omentum/metabolism , Peritoneal Dialysis , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Adult , Aged , Antibodies, Neutralizing/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Gene Expression Regulation , Humans , Interleukin-1beta/pharmacology , Male , Middle Aged , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/metabolism , Omentum/drug effects , Omentum/pathology , Primary Cell Culture , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Semaphorin-3A/pharmacology , Signal Transduction , Transforming Growth Factor beta1/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...