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.
Cancer Res ; 75(18): 3970-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26208903

ABSTRACT

Future curative cancer chemotherapies have to overcome tumor cell heterogeneity and plasticity. To test the hypothesis that the tumor suppressor maspin may reduce microenvironment-dependent prostate tumor cell plasticity and thereby modulate drug sensitivity, we established a new schematic combination of two-dimensional (2D), three-dimensional (3D), and suspension cultures to enrich prostate cancer cell subpopulations with distinct differentiation potentials. We report here that depending on the level of maspin expression, tumor cells in suspension and 3D collagen I manifest the phenotypes of stem-like and dormant tumor cell populations, respectively. In suspension, the surviving maspin-expressing tumor cells lost the self-renewal capacity, underwent senescence, lost the ability to dedifferentiate in vitro, and failed to generate tumors in vivo. Maspin-nonexpressing tumor cells that survived the suspension culture in compact tumorspheres displayed a higher level of stem cell marker expression, maintained the self-renewal capacity, formed tumorspheres in 3D matrices in vitro, and were tumorigenic in vivo. The drug sensitivities of the distinct cell subpopulations depend on the drug target and the differentiation state of the cells. In 2D, docetaxel, MS275, and salinomycin were all cytotoxic. In suspension, while MS275 and salinomycin were toxic, docetaxel showed no effect. Interestingly, cells adapted to 3D collagen I were only responsive to salinomycin. Maspin expression correlated with higher sensitivity to MS275 in both 2D and suspension and to salinomycin in 2D and 3D collagen I. Our data suggest that maspin reduces prostate tumor cell plasticity and enhances tumor sensitivity to salinomycin, which may hold promise in overcoming tumor cell heterogeneity and plasticity.


Subject(s)
Adenocarcinoma/metabolism , Drug Resistance, Neoplasm/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Serpins/physiology , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Adhesion/physiology , Cell Culture Techniques , Cell Dedifferentiation/physiology , Cell Line, Tumor , Cell Plasticity/drug effects , Cell Plasticity/physiology , Cell Self Renewal/physiology , Cellular Senescence , Docetaxel , Gene Expression Profiling , Heterografts , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Phenotype , Prostatic Neoplasms/pathology , Pyrans/pharmacology , Pyridines/pharmacology , Suspensions , Taxoids/pharmacology , Tumor Microenvironment
2.
Oncotarget ; 5(22): 11225-36, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25373490

ABSTRACT

The goal of the current study is to examine the biological effects of epithelial-specific tumor suppressor maspin on tumor host immune response. Accumulated evidence demonstrates an anti-tumor effect of maspin on tumor growth, invasion and metastasis. The molecular mechanism underlying these biological functions of maspin is thought to be through histone deacetylase inhibition, key to the maintenance of differentiated epithelial phenotype. Since tumor-driven stromal reactivities co-evolve in tumor progression and metastasis, it is not surprising that maspin expression in tumor cells inhibits extracellular matrix degradation, increases fibrosis and blocks hypoxia-induced angiogenesis. Using the athymic nude mouse model capable of supporting the growth and progression of xenogeneic human prostate cancer cells, we further demonstrate that maspin expression in tumor cells elicits neutrophil- and B cells-dependent host tumor immunogenicity. Specifically, mice bearing maspin-expressing tumors exhibited increased systemic and intratumoral neutrophil maturation, activation and antibody-dependent cytotoxicity, and decreased peritumoral lymphangiogenesis. These results reveal a novel biological function of maspin in directing host immunity towards tumor elimination that helps explain the significant reduction of xenograft tumor incidence in vivo and the clinical correlation of maspin with better prognosis of several types of cancer. Taken together, our data raised the possibility for novel maspin-based cancer immunotherapies.


Subject(s)
Prostatic Neoplasms/immunology , Serpins/immunology , Animals , Cell Differentiation/immunology , Cell Line, Tumor , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serpins/biosynthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...