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 ; 9(1): 3015, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816188

ABSTRACT

Tuberous Sclerosis Complex (TSC) and Lymphangioleiomyomatosis (LAM) are caused by inactivating mutations in TSC1 or TSC2, leading to mTORC1 hyperactivation. The mTORC1 inhibitors rapamycin and analogs (rapalogs) are approved for treating of TSC and LAM. Due to their cytostatic and not cytocidal action, discontinuation of treatment leads to tumor regrowth and decline in pulmonary function. Therefore, life-long rapalog treatment is proposed for the control of TSC and LAM lesions, which increases the chances for the development of acquired drug resistance. Understanding the signaling perturbations leading to rapalog resistance is critical for the development of better therapeutic strategies. We developed the first Tsc2-null rapamycin-resistant cell line, ELT3-245, which is highly tumorigenic in mice, and refractory to rapamycin treatment. In vitro ELT3-245 cells exhibit enhanced anchorage-independent cell survival, resistance to anoikis, and loss of epithelial markers. A key alteration in ELT3-245 is increased ß-catenin signaling. We propose that a subset of cells in TSC and LAM lesions have additional signaling aberrations, thus possess the potential to become resistant to rapalogs. Alternatively, when challenged with rapalogs TSC-null cells are reprogrammed to express mesenchymal-like markers. These signaling changes could be further exploited to induce clinically-relevant long-term remissions.


Subject(s)
Drug Resistance/genetics , Mesoderm/metabolism , Tuberous Sclerosis/genetics , Animals , Anoikis/drug effects , Anoikis/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/genetics , Drug Resistance/drug effects , Humans , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mesoderm/drug effects , Mice , Mutation/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology
2.
Cell Cycle ; 14(3): 399-407, 2015.
Article in English | MEDLINE | ID: mdl-25565629

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) increases translation, cell size and angiogenesis, and inhibits autophagy. mTORC1 is negatively regulated by hamartin and tuberin, the protein products of the tumor suppressors TSC1 and TSC2 that are mutated in Tuberous Sclerosis Complex (TSC) and sporadic Lymphangioleiomyomatosis (LAM). Hamartin interacts with the centrosomal and mitotic kinase polo-like kinase 1 (PLK1). Hamartin and tuberin deficient cells have abnormalities in centrosome duplication, mitotic progression, and cytokinesis, suggesting that the hamartin/tuberin heterodimer and mTORC1 signaling are involved in centrosome biology and mitosis. Here we report that PLK1 protein levels are increased in hamartin and tuberin deficient cells and LAM patient-derived specimens, and that this increase is rapamycin-sensitive. Pharmacological inhibition of PLK1 by the small-molecule inhibitor BI-2536 significantly decreased the viability and clonogenic survival of hamartin and tuberin deficient cells, which was associated with increased apoptosis. BI-2536 increased p62, LC3B-I and GFP-LC3 punctae, and inhibited HBSS-induced degradation of p62, suggesting that PLK1 inhibition attenuates autophagy. Finally, PLK1 inhibition repressed the expression and protein levels of key autophagy genes and proteins and the protein levels of Bcl(-)2 family members, suggesting that PLK1 regulates both autophagic and apoptotic responses. Taken together, our data point toward a previously unrecognized role of PLK1 on the survival of cells with mTORC1 hyperactivation, and the potential use of PLK1 inhibitors as novel therapeutics for tumors with dysregulated mTORC1 signaling, including TSC and LAM.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis/genetics , Autophagy/genetics , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Clone Cells , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Sirolimus/pharmacology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Polo-Like Kinase 1
3.
FASEB J ; 27(2): 581-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23104982

ABSTRACT

Fibroblast activation protein (FAP) is a serine protease selectively expressed on tumor stromal fibroblasts in epithelial carcinomas and is important in cancer growth, adhesion, and metastases. As FAP enzymatic activity is a potent therapeutic target, we aimed to identify inhibitory antibodies. Using a competitive inhibition strategy, we used phage display techniques to identify 53 single-chain variable fragments (scFvs) after three rounds of panning against FAP. These scFvs were expressed and characterized for binding to FAP by surface plasmon resonance and flow cytometry. Functional assessment of these antibodies yielded an inhibitory scFv antibody, named E3, which could attenuate 35% of FAP cleavage of the fluorescent substrate Ala-Pro-7-amido-4-trifluoromethylcoumarin compared with nonfunctional scFv control. Furthermore, a mutant E3 scFv was identified by yeast affinity maturation. It had higher affinity (4-fold) and enhanced inhibitory effect on FAP enzyme activity (3-fold) than E3. The application of both inhibitory anti-FAP scFvs significantly affected the formation of 3-dimensional FAP-positive cell matrix, as demonstrated by reducing the fibronectin fiber orientation from 41.18% (negative antibody control) to 34.06% (E3) and 36.15% (mutant E3), respectively. Thus, we have identified and affinity-maturated the first scFv antibody capable of inhibiting FAP function. This scFv antibody has the potential to disrupt the role of FAP in tumor invasion and metastasis.


Subject(s)
Gelatinases/antagonists & inhibitors , Gelatinases/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Serine Endopeptidases/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Affinity , Endopeptidases , Flow Cytometry , Gelatinases/genetics , Gelatinases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Targeted Therapy , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/immunology , Neoplasms/drug therapy , Neoplasms/enzymology , Peptide Library , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Surface Plasmon Resonance
4.
BMC Cancer ; 11: 245, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21668992

ABSTRACT

BACKGROUND: Alterations towards a permissive stromal microenvironment provide important cues for tumor growth, invasion, and metastasis. In this study, Fibroblast activation protein (FAP), a serine protease selectively produced by tumor-associated fibroblasts in over 90% of epithelial tumors, was used as a platform for studying tumor-stromal interactions. We tested the hypothesis that FAP enzymatic activity locally modifies stromal ECM (extracellular matrix) components thus facilitating the formation of a permissive microenvironment promoting tumor invasion in human pancreatic cancer. METHODS: We generated a tetracycline-inducible FAP overexpressing fibroblastic cell line to synthesize an in vivo-like 3-dimensional (3D) matrix system which was utilized as a stromal landscape for studying matrix-induced cancer cell behaviors. A FAP-dependent topographical and compositional alteration of the ECM was characterized by measuring the relative orientation angles of fibronectin fibers and by Western blot analyses. The role of FAP in the matrix-induced permissive tumor behavior was assessed in Panc-1 cells in assorted matrices by time-lapse acquisition assays. Also, FAP+ matrix-induced regulatory molecules in cancer cells were determined by Western blot analyses. RESULTS: We observed that FAP remodels the ECM through modulating protein levels, as well as through increasing levels of fibronectin and collagen fiber organization. FAP-dependent architectural/compositional alterations of the ECM promote tumor invasion along characteristic parallel fiber orientations, as demonstrated by enhanced directionality and velocity of pancreatic cancer cells on FAP+ matrices. This phenotype can be reversed by inhibition of FAP enzymatic activity during matrix production resulting in the disorganization of the ECM and impeded tumor invasion. We also report that the FAP+ matrix-induced tumor invasion phenotype is ß1-integrin/FAK mediated. CONCLUSION: Cancer cell invasiveness can be affected by alterations in the tumor microenvironment. Disruption of FAP activity and ß1-integrins may abrogate the invasive capabilities of pancreatic and other tumors by disrupting the FAP-directed organization of stromal ECM and blocking ß1-integrin dependent cell-matrix interactions. This provides a novel preclinical rationale for therapeutics aimed at interfering with the architectural organization of tumor-associated ECM. Better understanding of the stromal influences that fuel progressive tumorigenic behaviors may allow the effective future use of targeted therapeutics aimed at disrupting specific tumor-stromal interactions.


Subject(s)
Adenocarcinoma/pathology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/physiology , Fibroblasts/enzymology , Gelatinases/physiology , Membrane Proteins/physiology , Neoplasm Invasiveness/pathology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , Serine Endopeptidases/physiology , Tumor Microenvironment/physiology , Adenocarcinoma/enzymology , Animals , Blotting, Western , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor/enzymology , Cell Line, Tumor/pathology , Cell Movement , Collagen Type I/metabolism , Endopeptidases , Extracellular Matrix/ultrastructure , Fibronectins/metabolism , Fibronectins/ultrastructure , Focal Adhesion Kinase 1/physiology , Gelatinases/genetics , Humans , Integrin beta1/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred ICR , Mice, SCID , NIH 3T3 Cells/enzymology , Pancreatic Neoplasms/enzymology , Recombinant Fusion Proteins/physiology , Serine Endopeptidases/genetics , Time-Lapse Imaging , Transplantation, Heterologous
5.
Front Biosci (Elite Ed) ; 2(3): 1154-63, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515787

ABSTRACT

Fibroblast activation protein (FAP) is a type II integral membrane glycoprotein belonging to the serine protease family. It is selectively expressed by tumor stromal fibroblasts and transiently in the fibroblasts of healing wounds. FAP has been shown to modulate growth, differentiation, adhesion, and metastasis of tumor cells. Despite the importance of FAP in cancer, the mechanisms that govern its expression have not been defined. In this study, we determined the transcription start site of the FAP gene and identified a 2-kb segment with promoter activity in cells expressing FAP. Truncation of this fragment revealed that the core promoter activity resided in a 245-bp fragment surrounding the transcription start site. Electrophoretic mobility shift assay showed that EGR1 binds to the FAP promoter. Mutation of the EGR1 site within this fragment significantly decreased the promoter activity of FAP and eliminated EGR1 binding. Down-regulation of EGR1 resulted in a significant reduction in endogenous FAP mRNA expression. These findings identify the basal transcriptional requirements of FAP gene expression and show EGR1 is an important regulator of FAP expression.


Subject(s)
Gelatinases/genetics , Membrane Proteins/genetics , Promoter Regions, Genetic , Serine Endopeptidases/genetics , Animals , Base Sequence , Cell Line , DNA , DNA Primers , Electrophoretic Mobility Shift Assay , Endopeptidases , Humans , Mice , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcription, Genetic
6.
BMC Cancer ; 9: 94, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19323811

ABSTRACT

BACKGROUND: Interactions between cancer cells and stroma are critical for growth and invasiveness of epithelial tumors. The biochemical mechanisms behind tumor-stromal interactions leading to increased invasiveness and metastasis are mostly unknown. The goal of this study was to analyze the direct effects of staged stroma-derived extracellular matrices on breast cancer cell behavior. METHODS: Early and late three-dimensional matrices were produced by NIH-3T3 and tumor-associated murine fibroblasts, respectively. After removing fibroblasts, extracted matrices were re-cultured with breast epithelial cells of assorted characteristics: MCF-10A (non-tumorigenic), MCF-7 (tumorigenic, non-invasive), and MDA-MB-231 (tumorigenic, invasive). Effects prompted by staged matrices on epithelial cell's growth, morphology and invasion were determined. Also, matrix-induced velocity, directionality and relative track orientation of invasive cells were assessed in the presence or absence of inhibitors of phosphoinositide-3 kinase (PI3K) and/or beta-1 integrin. RESULTS: We observed that assorted breast epithelial cells reacted differently to two-dimensional vs. staged, control (early) and tumor-associated (late), three-dimensional matrices. MCF-10A had a proliferative advantage on two-dimensional substrates while MCF-7 and MDA-MB-231 showed no difference. MCF-10A and MCF-7 formed morphologically distinguishable aggregates within three-dimensional matrices, while MDA-MB-231 exhibited increased spindle-shape morphologies and directional movements within three-dimensional matrices. Furthermore, MDA-MB-231 acquired a pattern of parallel oriented organization within tumor-associated, but not control matrices. Moreover, tumor-associated matrices induced PI3K and beta1-integrin dependent Akt/PKB activity in MDA-MB-231 cells. Interestingly, beta1-integrin (but not PI3K) regulated tumor-associated matrix-induced mesenchymal invasion which, when inhibited, resulted in a change of invasive strategy rather than impeding invasion altogether. CONCLUSION: We propose that both cells and matrices are important to promote effective breast cancer cell invasion through three-dimensional matrices and that beta1-integrin inhibition is not necessarily sufficient to block tumor-matrix induced breast cancer cell invasion. Additionally, we believe that characterizing stroma staging (e.g., early vs. late or tumor-associated) might be beneficial for predicting matrix-induced cancer cell responses in order to facilitate the selection of therapies.


Subject(s)
Extracellular Matrix/physiology , Integrin beta1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation , Cell Shape/physiology , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Integrin beta1/immunology , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology
7.
Gynecol Oncol ; 110(1): 99-109, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18448156

ABSTRACT

OBJECTIVE: Due to a lack of experimental systems, little is known about ovarian stroma. Here, we introduce an in vivo-like 3-D system of mesenchymal stromal progression during ovarian tumorigenesis to support the study of stroma permissiveness in human ovarian neoplasias. METHODS: To sort 3-D cultures into 'normal,' 'primed' and 'activated' stromagenic stages, 29 fibroblastic cell lines from 5 ovarian tumor samples (tumor ovarian fibroblasts, TOFs) and 14 cell lines from normal prophylactic oophorectomy samples (normal ovarian fibroblasts, NOFs) were harvested and characterized for their morphological, biochemical and 3-D culture features. RESULTS: Under 2-D conditions, cells displayed three distinct morphologies: spread, spindle, and intermediate. We found that spread and spindle cells have similar levels of alpha-SMA, a desmoplastic marker, and consistent ratios of pFAKY(397)/totalFAK. In 3-D intermediate cultures, alpha-SMA levels were virtually undetectable while pFAKY(397)/totalFAK ratios were low. In addition, we used confocal microscopy to assess in vivo-like extracellular matrix topography, nuclei morphology and alpha-SMA features in the 3-D cultures. We found that all NOFs presented 'normal' characteristics, while TOFs presented both 'primed' and 'activated' features. Moreover, immunohistochemistry analyses confirmed that the 3-D matrix-dependent characteristics are reminiscent of those observed in in vivo stromal counterparts. CONCLUSIONS: We conclude that primary human ovarian fibroblasts maintain in vivo-like (staged) stromal characteristics in a 3-D matrix-dependent manner. Therefore, our stromal 3-D system offers a tool that can enhance the understanding of both stromal progression and stroma-induced ovarian tumorigenesis. In the future, this system could also be used to develop ovarian stroma-targeted therapies.


Subject(s)
Fibroblasts/physiology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology , Ovary/physiology , Stromal Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Line, Tumor , Female , Fibroblasts/cytology , Fibroblasts/pathology , Humans , Mice , Neoplasm Staging , Ovary/cytology , Rabbits
8.
Mol Cancer Ther ; 4(3): 351-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767544

ABSTRACT

Tumor-associated fibroblasts are functionally and phenotypically distinct from normal fibroblasts that are not in the tumor microenvironment. Fibroblast activation protein is a 95 kDa cell surface glycoprotein expressed by tumor stromal fibroblasts, and has been shown to have dipeptidyl peptidase and collagenase activity. Site-directed mutagenesis at the catalytic site of fibroblast activation protein, Ser624 --> Ala624, resulted in an approximately 100,000-fold loss of fibroblast activation protein dipeptidyl peptidase (DPP) activity. HEK293 cells transfected with wild-type fibroblast activation protein, enzymatic mutant (S624A) fibroblast activation protein, or vector alone, were inoculated subcutaneously into immunodeficient mouse to assess the contribution of fibroblast activation protein enzymatic activity to tumor growth. Overexpression of wild-type fibroblast activation protein showed growth potentiation and enhanced tumorigenicity compared with both fibroblast activation protein S624A and vector-transfected HEK293 xenografts. HEK293 cells transfected with fibroblast activation protein S624A showed tumor growth rates and tumorigenicity potential similar only to vector-transfected HEK293. In vivo assessment of fibroblast activation protein DPP activity of these tumors showed enhanced enzymatic activity of wild-type fibroblast activation protein, with only baseline levels of fibroblast activation protein DPP activity in either fibroblast activation protein S624A or vector-only xenografts. These results indicate that the enzymatic activity of fibroblast activation protein is necessary for fibroblast activation protein-driven tumor growth in the HEK293 xenograft model system. This establishes the proof-of-principle that the enzymatic activity of fibroblast activation protein plays an important role in the promotion of tumor growth, and provides an attractive target for therapeutics designed to alter fibroblast activation protein-induced tumor growth by targeting its enzymatic activity.


Subject(s)
Antigens, Neoplasm/physiology , Biomarkers, Tumor/physiology , Fibroblasts/metabolism , Neoplasms/enzymology , Serine Endopeptidases/physiology , Alanine/chemistry , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Biomarkers, Tumor/metabolism , Blotting, Western , Catalytic Domain , Cell Line , Cell Line, Tumor , Cell Proliferation , DNA, Complementary/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Endopeptidases , Flow Cytometry , Gelatinases , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Kinetics , Membrane Proteins , Mice , Microscopy, Fluorescence , Models, Molecular , Neoplasm Transplantation , Serine/chemistry , Serine Endopeptidases/metabolism , Time Factors , Transfection
9.
Cancer Res ; 62(16): 4767-72, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12183436

ABSTRACT

Fibroblast activation protein (FAP) is a type II integral membrane glycoprotein belonging to the serine protease family. Human FAP is selectively expressed by tumor stromal fibroblasts in epithelial carcinomas, but not by epithelial carcinoma cells, normal fibroblasts, or other normal tissues. FAP has been shown to have both in vitro dipeptidyl peptidase and collagenase activity, but its biological function in the tumor microenvironment is unknown. The modeled structure of murine FAP consists of a short cytoplasmic tail, a single hydrophobic transmembrane region, and a large extracellular domain. A seven-bladed beta-propeller domain is situated on top of the catalytic triad and may serve as a "gate" to selectively filter protein access to the catalytic site. HEK293 cells transfected to constitutively express murine FAP, when xenografted into scid mice, were 2-4 times more likely to develop s.c. tumors and showed a 10-40-fold enhancement of tumor growth compared with mock-transfected HEK293 cells. Rabbits immunized with recombinant murine FAP developed polyclonal anti-FAP antibodies that significantly inhibited murine FAP dipeptidyl peptidase activity in vitro. HT-29 xenografts treated with these inhibitory anti-FAP antisera exhibited attenuated growth compared with tumors treated with preimmunization rabbit antisera. These data demonstrate the ability of FAP to potentiate tumor growth in an animal model. Moreover, tumor growth is attenuated by antibodies that inhibit the proteolytic activity of FAP. These findings suggest a possible therapeutic role for functional inhibition of FAP activity.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Growth Substances/physiology , Serine Endopeptidases/physiology , Animals , Antibodies/pharmacology , Cell Division/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Disease Models, Animal , Endopeptidases , Gelatinases , Growth Substances/chemistry , Growth Substances/immunology , HT29 Cells/pathology , Humans , Membrane Proteins , Mice , Mice, SCID , Protein Conformation , Protein Structure, Tertiary , Serine Endopeptidases/chemistry , Serine Endopeptidases/immunology , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...