Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7(1): 14167, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29074857

ABSTRACT

The use of in vitro, engineered surrogates in the field of cancer research is of interest for studies involving mechanisms of growth and metastasis, and response to therapeutic intervention. While biomimetic surrogates better model human disease, their complex composition and dimensionality make them challenging to evaluate in a real-time manner. This feature has hindered the broad implementation of these models, particularly in drug discovery. Herein, several methods and approaches for the real-time, non-invasive analysis of cell growth and response to treatment in tissue-engineered, three-dimensional models of breast cancer are presented. The tissue-engineered surrogates used to demonstrate these methods consist of breast cancer epithelial cells and fibroblasts within a three dimensional volume of extracellular matrix and are continuously perfused with nutrients via a bioreactor system. Growth of the surrogates over time was measured using optical in vivo (IVIS) imaging. Morphologic changes in specific cell populations were evaluated by multi-photon confocal microscopy. Response of the surrogates to treatment with paclitaxel was measured by optical imaging and by analysis of lactate dehydrogenase and caspase-cleaved cytokeratin 18 in the perfused medium. Each method described can be repeatedly performed during culture, allowing for real-time, longitudinal analysis of cell populations within engineered tumor models.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Tissue Engineering/instrumentation , Tissue Engineering/methods , Animals , Bioreactors , Breast Neoplasms/drug therapy , Cell Proliferation , Cell Survival/drug effects , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Equipment Design , Extracellular Matrix/pathology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Keratin-18/metabolism , L-Lactate Dehydrogenase/metabolism , Luminescent Measurements/methods , Mice , Microscopy, Confocal , Paclitaxel/pharmacology , Xenograft Model Antitumor Assays
2.
Cancer Biol Ther ; 8(19): 1840-51, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19717984

ABSTRACT

The transcription factors Notch1 and KLF4 specify epithelial cell fates and confer stem cell properties. Suggesting a functional relationship, each gene can act to promote or suppress tumorigenesis in a context-dependent manner, and alteration of KLF4 or Notch pathway genes in mice gives rise to similar phenotypes. Activation of a conditional allele of KLF4 in RK3E epithelial cells rapidly induces expression of Notch1 mRNA and the active, intracellular form of Notch1. KLF4-induced transformation was suppressed by knockdown of endogenous Notch1 using siRNA or an inhibitor of gamma-secretase. Chromatin immunoprecipitation assay shows that KLF4 binds to the proximal Notch1 promoter in human mammary epithelial cells, and siRNA-mediated suppression of KLF4 in human mammary cancer cells results in reduced expression of Notch1. Furthermore, KLF4 and Notch1 expression are correlated in primary human breast tumors (N = 89; Pearson analysis, r > 0.5, p < 0.0001). Like KLF4, Notch1 was previously shown to induce transformation of rat cells immortalized with adenovirus E1A, similar to RK3E cells. We therefore compared the signaling requirements for Notch1- or KLF4-induced malignant transformation of RK3E. As expected, transformation by Notch1 was suppressed by dominant-negative CSL or MAML1, inhibitors of canonical Notch1 signaling. However, these inhibitors did not suppress transformation by KLF4. Therefore, while KLF4-induced transformation requires Notch1, canonical Notch1 signaling is not required, and Notch1 may signal through a distinct pathway in cells with increased KLF4 activity. These results suggest that KLF4 could contribute to breast tumor progression by activating synthesis of Notch1 and by promoting signaling through a non-canonical Notch1 pathway.


Subject(s)
Cell Transformation, Neoplastic/genetics , Kruppel-Like Transcription Factors/genetics , Receptor, Notch1/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunoblotting , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Rats , Receptor, Notch1/biosynthesis , Receptor, Notch1/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Cancer Biol Ther ; 8(3): 289-98, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19197145

ABSTRACT

The transcription factor KLF4 acts in post-mitotic epithelial cells to promote differentiation and functions in a context-dependent fashion as an oncogene. In the skin KLF4 is co-expressed with the nuclear receptors RARgamma and RXRalpha, and formation of the skin permeability barrier is a shared function of these three proteins. We utilized a KLF4-transgenic mouse model of skin cancer in combination with cultured epithelial cells to examine functional interactions between KLF4 and retinoic acid receptors. In cultured cells, activation of a conditional, KLF4-estrogen receptor fusion protein by 4-hydroxytamoxifen resulted in rapid upregulation of transcripts for nuclear receptors including RARgamma and RXRalpha. We tested retinoids in epithelial cell transformation assays, including an RAR-selective agonist (all-trans RA), an RXR-selective agonist (9-cis UAB30, rexinoid), and a pan agonist (9-cis RA). Unlike for several other genes, transformation by KLF4 was inhibited by each retinoid, implicating distinct nuclear receptor heterodimers as modulators of KLF4 transforming activity. When RXRalpha expression was suppressed by RNAi in cultured cells, transformation was promoted and the inhibitory effect of 9-cis UAB30 was attenuated. Similarly as shown for other mouse models of skin cancer, rexinoid prevented skin tumor initiation resulting from induction of KLF4 in basal keratinocytes. Rexinoid permitted KLF4 expression and KLF4-induced cell cycling, but attenuated the KLF4-induced misexpression of cytokeratin 1 in basal cells. Neoplastic lesions including hyperplasia, dysplasia and squamous cell carcinoma-like lesions were prevented for up to 30 days. Taken together, the results identify retinoid receptors including RXRalpha as ligand-dependent inhibitors of KLF4-mediated transformation or tumorigenesis.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Kruppel-Like Transcription Factors/antagonists & inhibitors , Naphthalenes/pharmacology , Neoplasms, Squamous Cell/prevention & control , Skin Neoplasms/prevention & control , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line , Gene Expression Regulation, Neoplastic/drug effects , Humans , Keratins/biosynthesis , Keratins/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Mice , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , Rats , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Retinoid X Receptor alpha/agonists , Retinoid X Receptor alpha/biosynthesis , Retinoid X Receptor alpha/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tretinoin/pharmacology , Retinoic Acid Receptor gamma
5.
Cancer Biol Ther ; 5(6): 674-83, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16855373

ABSTRACT

The hedgehog pathway regulates epithelial-mesenchymal interactions, differentiation, proliferation and survival during development. Stimulation of hedgehog signaling induces carcinogenesis or promotes cell survival in cancers of multiple organs. Using real-time, quantitative PCR, laser capture microdissection, and immunohistochemistry, distinctive patterns of expression of the hedgehog pathway members patched 1 (PTCH1), smoothened, GLI1, GLI2 and the 3 hedgehog ligands were identified for epithelial cells and stromal fibroblasts in benign breast and breast cancer. Hedgehog ligands were expressed at higher levels in some cancer epithelial cell lines compared to noncancerous epithelial cells. Correspondingly, expression of GLI1, a transcription factor and transcriptional product of hedgehog signaling, was increased 8-fold in cancer epithelial cell lines; however, PTCH1, also a transcriptional target of hedgehog signaling in many cell types, was not increased. GLI1 protein and mRNA, and PTCH1 and sonic hedgehog (SHH) proteins were elevated in 3 of 10 breast cancers; however, PTCH1 transcripts were not consistently increased. Hedgehog-mediated transcription, as indicated by a reporter of GLI-dependent promoter activity and by expression of GLI1 transcripts, was reduced by the hedgehog pathway inhibitor cyclopamine in both MDA-MB-435 cancer epithelial cells and MCF10AT epithelial cells, a cell line derived from benign breast. However, cyclopamine reduced viability of cancer epithelial cell lines, including MDA-MB-435, but did not specifically affect fibroblasts or epithelial cells from benign breast, including MCF10AT. Treatment with sonic hedgehog ligand diminished the cyclopamine-induced reduction in GLI-dependent promoter activity in MCF10AT and MDA-MB-435 and viability of MDA-MB-435. These results demonstrate modulation of GLI-mediated transcription in both cancer and benign-derived epithelial cells by cyclopamine and sonic hedgehog, and further suggest that hedgehog signaling contributes to the survival of only the cancer epithelial cells. Determination as to whether the increase in GLI1 and SHH expression in breast cancer indicates a significant increase in hedgehog signaling will require further evaluation.


Subject(s)
Breast Neoplasms/physiopathology , Breast/physiology , Hedgehog Proteins/physiology , Veratrum Alkaloids/pharmacology , Base Sequence , Breast/drug effects , Cell Line, Tumor , DNA Primers , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Stromal Cells/drug effects , Stromal Cells/physiology
6.
Cancer Biol Ther ; 4(12): 1401-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16357510

ABSTRACT

KLF4 is induced upon growth-arrest in vitro and during epithelial maturation in vivo, and is essential for proper cell fate specification of post-mitotic cells. In spite of a normal role in post-mitotic cells, expression is upregulated and constitutive in certain tumor types. KLF4 functions as an oncogene in vitro, and enforced expression in basal cells of mouse skin rapidly induces lesions similar to hyperplasia, dysplasia and squamous cell carcinoma (SCC). Here we used conditional expression to characterize early steps in KLF4-mediated tumor initiation. In contrast to SCC-like lesions that result when using a conditional, keratin 14 promoter-dependent strategy, lower conditional expression achieved using a MMTV promoter induced only epidermal cycling within morphologically normal skin, a process we termed occult cell turnover. Surprisingly, KLF4-induced hyperplastic lesions showed increased transgene-derived mRNA and protein in maturing, PCNA-negative cells, a property of endogenous KLF4. In contrast, hyperplastic lesions induced by GLI1, a control, showed uniform transgene expression. In KLF4-induced dysplasia and SCC the complementarity of KLF4 and PCNA was replaced by concordance of the two proteins. These studies show that KLF4 transcripts are normally suppressed in cycling cells in a promoter-independent fashion, consistent with a post-transcriptional control, and reveal loss of this control in the transition from hyperplasia to dysplasia. Like the mouse tumors, human cutaneous SCCs and adjacent dysplasias frequently showed maturation-independence of KLF4, with co-expression of KLF4 and PCNA. A smaller subset of human SCCs showed complementarity of KLF4 and PCNA, similar to hyperplastic mouse skin. The results identify parallels between a mouse model and human primary tumors, and show that successive increases of KLF4 in the nuclei of basal keratinocytes leads to occult cell turnover followed by hyperplasia, dysplasia, and invasive SCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Transformation, Neoplastic/chemically induced , Kruppel-Like Transcription Factors/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Skin Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line , Disease Models, Animal , Disease Progression , Doxorubicin/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Transgenic , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transgenes , Zinc Finger Protein GLI1
7.
Oncogene ; 24(9): 1491-500, 2005 Feb 24.
Article in English | MEDLINE | ID: mdl-15674344

ABSTRACT

KLF4/GKLF normally functions in differentiating epithelial cells, but also acts as a transforming oncogene in vitro. To examine the role of this zinc finger protein in skin, we expressed the wild-type human allele from inducible and constitutive promoters. When induced in basal keratinocytes, KLF4 rapidly abolished the distinctive properties of basal and parabasal epithelial cells. KLF4 caused a transitory apoptotic response and the skin progressed through phases of hyperplasia and dysplasia. By 6 weeks, lesions exhibited nuclear KLF4 and other morphologic and molecular similarities to squamous cell carcinoma in situ. p53 determined the patch size sufficient to establish lesions, as induction in a mosaic pattern produced skin lesions only when p53 was deficient. Compared with p53 wild-type animals, p53 hemizygous animals had early onset of lesions and a pronounced fibrovascular response that included outgrowth of subcutaneous sarcoma. A KLF4-estrogen receptor fusion protein showed tamoxifen-dependent nuclear localization and conditional transformation in vitro. The results suggest that KLF4 can function in the nucleus to induce squamous epithelial dysplasia, and indicate roles for p53 and epithelial-mesenchymal signaling in these early neoplastic lesions.


Subject(s)
Cell Differentiation/physiology , Cell Division/drug effects , DNA-Binding Proteins/biosynthesis , Epithelial Cells/pathology , Keratinocytes/cytology , Skin/pathology , Transcription Factors/biosynthesis , Animals , Apoptosis/drug effects , Crosses, Genetic , DNA Primers , Doxorubicin/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Transfection
8.
Clin Cancer Res ; 10(8): 2709-19, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15102675

ABSTRACT

PURPOSE: The Krüppel-like transcription factor KLF4/GKLF induces both malignant transformation and a slow-growth phenotype in vitro. Although KLF4 expression is increased in most cases of breast cancer, it was unknown whether these cases represent a distinct subtype with a different clinical outcome. EXPERIMENTAL DESIGN: We examined expression of KLF4 by immunostaining 146 cases of human primary infiltrating ductal carcinoma of the breast. Staining patterns were correlated with clinical outcome and with established prognostic factors. RESULTS: Subcellular localization exhibited case-to-case variation. Tumors with high nuclear staining and low cytoplasmic staining were termed type 1. For patients with early-stage disease (i.e., stage I or IIA), type 1 staining was associated with eventual death because of breast cancer (hazard ratio, 2.8; 95% confidence interval, 1.23-6.58; P = 0.011). The association was stronger in patients with early-stage cancer and small primary tumors (i.e., < or =2.0 cm in diameter; hazard ratio, 4.3; 95% confidence interval, 1.75-10.62; P < 0.001). For patients with early-stage disease, multivariate analysis indicated that type 1 staining was independently associated with outcome (adjusted hazard ratio 2.6; 95% confidence interval, 1.10-6.05; P = 0.029). Type 1 staining was also associated with high histological grade (P = 0.032), increased expression of Ki67 (P = 0.016), and reduced expression of BCL2 (P = 0.032). In vitro, KLF4 was localized within the nucleus of transformed RK3E epithelial cells, consistent with a nuclear function of this transcription factor during induction of malignant transformation. CONCLUSIONS: The results suggest that localization of KLF4 in the nucleus of breast cancer cells is a prognostic factor and identify KLF4 as a marker of an aggressive phenotype in early-stage infiltrating ductal carcinoma.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Nucleus/metabolism , DNA-Binding Proteins/biosynthesis , Transcription Factors/biosynthesis , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic , Cytoplasm/metabolism , Cytosol/metabolism , DNA, Complementary/metabolism , Disease-Free Survival , Epitopes/chemistry , Female , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Multivariate Analysis , Phenotype , Plasmids/metabolism , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Regression Analysis , Time Factors , Transfection , Treatment Outcome , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...